메뉴 건너뛰기




Volumn 6, Issue 22, 2015, Pages 18748-18779

Targeting cancer by binding iron: Dissecting cellular signaling pathways

Author keywords

Cancer; Chelators; Iron; Signaling; Thiosemicarbazones

Indexed keywords

3 AMINOPICOLINALDEHYDE THIOSEMICARBAZONE; CARBOPLATIN; CYCLIN DEPENDENT KINASE; CYCLIN DEPENDENT KINASE INHIBITOR; CYCLINE; CYCLOPHOSPHAMIDE; DEFERASIROX; DEFEROXAMINE MESYLATE; DI 2 PYRIDYLKETONE 2 METHYL 3 THIOSEMICARBAZONE; DI 2 PYRIDYLKETONE 4,4 DIMETHYL 3 THIOSEMICARBAZONE; DI 2 PYRIDYLKETONE THIOSEMICARBAZONE; ETOPOSIDE; GELATINASE B; HYDRAZONE DERIVATIVE; IRON CHELATING AGENT; MAMMALIAN TARGET OF RAPAMYCIN; MITOGEN ACTIVATED PROTEIN KINASE; MYC PROTEIN; NERVE CELL ADHESION MOLECULE; OCCLUDIN; PROTEIN KINASE B; RAF PROTEIN; RAS PROTEIN; RIBONUCLEOTIDE REDUCTASE; STAT3 PROTEIN; STRESS ACTIVATED PROTEIN KINASE; SYNAPTOPHYSIN; TRANSFORMING GROWTH FACTOR BETA; UNCLASSIFIED DRUG; UNINDEXED DRUG; WNT PROTEIN; ANTINEOPLASTIC AGENT; IRON; THIOSEMICARBAZONE DERIVATIVE;

EID: 84938833826     PISSN: None     EISSN: 19492553     Source Type: Journal    
DOI: 10.18632/oncotarget.4349     Document Type: Article
Times cited : (142)

References (318)
  • 4
    • 29844457587 scopus 로고    scopus 로고
    • The evolution of iron chelators for the treatment of iron overload disease and cancer
    • Kalinowski DS, Richardson DR. The evolution of iron chelators for the treatment of iron overload disease and cancer. Pharmacol Rev. 2005; 57:547-583.
    • (2005) Pharmacol Rev. , vol.57 , pp. 547-583
    • Kalinowski, D.S.1    Richardson, D.R.2
  • 7
    • 79952227712 scopus 로고    scopus 로고
    • Ironing out cancer
    • Torti SV, Torti FM. Ironing out cancer. Cancer Res. 2011; 71:1511-1514.
    • (2011) Cancer Res. , vol.71 , pp. 1511-1514
    • Torti, S.V.1    Torti, F.M.2
  • 10
    • 4243607831 scopus 로고
    • Toxicity of iron and hydrogen peroxide: the Fenton reaction
    • Winterbourn CC. Toxicity of iron and hydrogen peroxide: the Fenton reaction. Toxicol Lett. 1995; 82-83:969-974.
    • (1995) Toxicol Lett , vol.82-83 , pp. 969-974
    • Winterbourn, C.C.1
  • 11
    • 0031567095 scopus 로고    scopus 로고
    • The molecular mechanisms of the metabolism and transport of iron in normal and neoplastic cells
    • Richardson DR, Ponka P. The molecular mechanisms of the metabolism and transport of iron in normal and neoplastic cells. Biochim Biophys Acta. 1997; 1331:1-40.
    • (1997) Biochim Biophys Acta. , vol.1331 , pp. 1-40
    • Richardson, D.R.1    Ponka, P.2
  • 13
    • 84927923911 scopus 로고    scopus 로고
    • Duodenal Cytochrome b (DCYTB) in Iron Metabolism: An Update on Function and Regulation
    • Lane DJ, Bae DH, Merlot AM, Sahni S, Richardson DR. Duodenal Cytochrome b (DCYTB) in Iron Metabolism: An Update on Function and Regulation. Nutrients. 2015; 7:2274-2296.
    • (2015) Nutrients. , vol.7 , pp. 2274-2296
    • Lane, D.J.1    Bae, D.H.2    Merlot, A.M.3    Sahni, S.4    Richardson, D.R.5
  • 14
    • 0019860059 scopus 로고
    • Transferrin, biochemistry, physiology and clinical significance
    • Morgan EH. Transferrin, biochemistry, physiology and clinical significance. Mol Aspects Med. 1981; 4:1-123.
    • (1981) Mol Aspects Med. , vol.4 , pp. 1-123
    • Morgan, E.H.1
  • 15
    • 0031963449 scopus 로고    scopus 로고
    • Function and regulation of transferrin and ferritin
    • Ponka P, Beaumont C, Richardson DR. Function and regulation of transferrin and ferritin. Semin Hematol. 1998; 35:35-54.
    • (1998) Semin Hematol. , vol.35 , pp. 35-54
    • Ponka, P.1    Beaumont, C.2    Richardson, D.R.3
  • 17
    • 8944224496 scopus 로고
    • Transferrin: Receptor-Mediated Endocytosis and Iron Delivery
    • Pastan I, Willingham M, eds, New York: Springer US
    • Hanover JA, Dickson RB. (1985). Transferrin: Receptor-Mediated Endocytosis and Iron Delivery. In: Pastan I, Willingham M, eds. Endocytosis. (New York: Springer US), pp. 131-161.
    • (1985) Endocytosis , pp. 131-161
    • Hanover, J.A.1    Dickson, R.B.2
  • 20
    • 0037108199 scopus 로고    scopus 로고
    • The labile iron pool: characterization, measurement, and participation in cellular processes
    • Kakhlon O, Cabantchik ZI. The labile iron pool: characterization, measurement, and participation in cellular processes. Free Radic Biol Med. 2002; 33:1037-1046.
    • (2002) Free Radic Biol Med. , vol.33 , pp. 1037-1046
    • Kakhlon, O.1    Cabantchik, Z.I.2
  • 21
    • 0017700831 scopus 로고
    • Low molecular weight intracellular iron transport compounds
    • Jacobs A. Low molecular weight intracellular iron transport compounds. Blood. 1977; 50:433-439.
    • (1977) Blood. , vol.50 , pp. 433-439
    • Jacobs, A.1
  • 22
    • 0029865751 scopus 로고    scopus 로고
    • Distribution of iron in reticulocytes after inhibition of heme synthesis with succinylacetone: examination of the intermediates involved in iron metabolism
    • Richardson DR, Ponka P, Vyoral D. Distribution of iron in reticulocytes after inhibition of heme synthesis with succinylacetone: examination of the intermediates involved in iron metabolism. Blood. 1996; 87:3477-3488.
    • (1996) Blood. , vol.87 , pp. 3477-3488
    • Richardson, D.R.1    Ponka, P.2    Vyoral, D.3
  • 27
    • 0032410688 scopus 로고    scopus 로고
    • Iron regulatory proteins, iron responsive elements and iron homeostasis
    • Eisenstein RS, Blemings KP. Iron regulatory proteins, iron responsive elements and iron homeostasis. J Nutr. 1998; 128:2295-2298.
    • (1998) J Nutr. , vol.128 , pp. 2295-2298
    • Eisenstein, R.S.1    Blemings, K.P.2
  • 28
    • 0029758487 scopus 로고    scopus 로고
    • Molecular control of vertebrate iron metabolism: mRNA-based regulatory circuits operated by iron, nitric oxide, and oxidative stress
    • Hentze MW, Kuhn LC. Molecular control of vertebrate iron metabolism: mRNA-based regulatory circuits operated by iron, nitric oxide, and oxidative stress. Proc Natl Acad Sci U S A. 1996; 93:8175-8182.
    • (1996) Proc Natl Acad Sci U S A. , vol.93 , pp. 8175-8182
    • Hentze, M.W.1    Kuhn, L.C.2
  • 29
    • 84876854791 scopus 로고    scopus 로고
    • Iron and cancer: more ore to be mined
    • Torti SV, Torti FM. Iron and cancer: more ore to be mined. Nat Rev Cancer. 2013; 13:342-355.
    • (2013) Nat Rev Cancer. , vol.13 , pp. 342-355
    • Torti, S.V.1    Torti, F.M.2
  • 30
    • 0022475665 scopus 로고
    • Transferrin receptor expression in non-malignant and malignant human breast tissue
    • Walker RA, Day SJ. Transferrin receptor expression in non-malignant and malignant human breast tissue. J Pathol. 1986; 148:217-224.
    • (1986) J Pathol. , vol.148 , pp. 217-224
    • Walker, R.A.1    Day, S.J.2
  • 32
    • 0023153556 scopus 로고
    • Transferrin receptor expression in normal skin and in various cutaneous tumors
    • Soyer HP, Smolle J, Torne R, Kerl H. Transferrin receptor expression in normal skin and in various cutaneous tumors. J Cutan Path. 1987; 14:1-5.
    • (1987) J Cutan Path. , vol.14 , pp. 1-5
    • Soyer, H.P.1    Smolle, J.2    Torne, R.3    Kerl, H.4
  • 33
    • 0025341256 scopus 로고
    • The uptake of iron and transferrin by the human malignant melanoma cell
    • Richardson DR, Baker E. The uptake of iron and transferrin by the human malignant melanoma cell. Biochim Biophys Acta. 1990; 1053:1-12.
    • (1990) Biochim Biophys Acta. , vol.1053 , pp. 1-12
    • Richardson, D.R.1    Baker, E.2
  • 34
    • 0022616986 scopus 로고
    • Mechanisms of growth inhibition by anti-transferrin receptor monoclonal antibodies
    • Taetle R, Castagnola J, Mendelsohn J. Mechanisms of growth inhibition by anti-transferrin receptor monoclonal antibodies. Cancer Res. 1986; 46:1759-1763.
    • (1986) Cancer Res. , vol.46 , pp. 1759-1763
    • Taetle, R.1    Castagnola, J.2    Mendelsohn, J.3
  • 35
    • 0019459845 scopus 로고
    • Role of ribonucleotide reductase in expression in the neoplastic program
    • Takeda E, Weber G. Role of ribonucleotide reductase in expression in the neoplastic program. Life Sci. 1981; 28:1007-1014.
    • (1981) Life Sci. , vol.28 , pp. 1007-1014
    • Takeda, E.1    Weber, G.2
  • 38
    • 77951037271 scopus 로고    scopus 로고
    • Manipulation of iron transporter genes results in the suppression of human and mouse mammary adenocarcinomas
    • Jiang XP, Elliott RL, Head JF. Manipulation of iron transporter genes results in the suppression of human and mouse mammary adenocarcinomas. Anticancer Res. 2010; 30:759-765.
    • (2010) Anticancer Res. , vol.30 , pp. 759-765
    • Jiang, X.P.1    Elliott, R.L.2    Head, J.F.3
  • 42
    • 0021128761 scopus 로고
    • Iron depletion: possible cause of tumor cell cytotoxicity induced by activated macrophages
    • Hibbs JB, Jr., Taintor RR, Vavrin Z. Iron depletion: possible cause of tumor cell cytotoxicity induced by activated macrophages. Biochem Biophys Res Commun. 1984; 123:716-723.
    • (1984) Biochem Biophys Res Commun. , vol.123 , pp. 716-723
    • Hibbs, J.B.1    Taintor, R.R.2    Vavrin, Z.3
  • 44
    • 0025883342 scopus 로고
    • Nitric oxide: physiology, pathophysiology, and pharmacology
    • Moncada S, Palmer RM, Higgs EA. Nitric oxide: physiology, pathophysiology, and pharmacology. Pharmacol Rev. 1991; 43:109-142.
    • (1991) Pharmacol Rev. , vol.43 , pp. 109-142
    • Moncada, S.1    Palmer, R.M.2    Higgs, E.A.3
  • 45
    • 0028784211 scopus 로고
    • The effect of redox-related species of nitrogen monoxide on transferrin and iron uptake and cellular proliferation of erythroleukemia (K562) cells
    • Richardson DR, Neumannova V, Nagy E, Ponka P. The effect of redox-related species of nitrogen monoxide on transferrin and iron uptake and cellular proliferation of erythroleukemia (K562) cells. Blood. 1995; 86:3211-3219.
    • (1995) Blood. , vol.86 , pp. 3211-3219
    • Richardson, D.R.1    Neumannova, V.2    Nagy, E.3    Ponka, P.4
  • 46
    • 0034646393 scopus 로고    scopus 로고
    • Nitrogen monoxide activates iron regulatory protein 1 RNA-binding activity by two possible mechanisms: effect on the [4Fe-4S] cluster and iron mobilization from cells
    • Wardrop SL, Watts RN, Richardson DR. Nitrogen monoxide activates iron regulatory protein 1 RNA-binding activity by two possible mechanisms: effect on the [4Fe-4S] cluster and iron mobilization from cells. Biochemistry 2000; 39:2748-2758.
    • (2000) Biochemistry , vol.39 , pp. 2748-2758
    • Wardrop, S.L.1    Watts, R.N.2    Richardson, D.R.3
  • 47
    • 0029030330 scopus 로고
    • Nitrogen monoxide decreases iron uptake from transferrin but does not mobilise iron from prelabelled neoplastic cells
    • Richardson DR, Neumannova V, Ponka P. Nitrogen monoxide decreases iron uptake from transferrin but does not mobilise iron from prelabelled neoplastic cells. Biochim Biophys Acta. 1995; 1266:250-260.
    • (1995) Biochim Biophys Acta. , vol.1266 , pp. 250-260
    • Richardson, D.R.1    Neumannova, V.2    Ponka, P.3
  • 48
    • 0035895946 scopus 로고    scopus 로고
    • Nitrogen monoxide (NO) and glucose: unexpected links between energy metabolism and NO-mediated iron mobilization from cells
    • Watts RN, Richardson DR. Nitrogen monoxide (NO) and glucose: unexpected links between energy metabolism and NO-mediated iron mobilization from cells. J Biol Chem. 2001; 276:4724-4732.
    • (2001) J Biol Chem. , vol.276 , pp. 4724-4732
    • Watts, R.N.1    Richardson, D.R.2
  • 49
    • 84855271366 scopus 로고    scopus 로고
    • Nitric oxide storage and transport in cells are mediated by glutathione S-transferase P1-1 and multidrug resistance protein 1 via dinitrosyl iron complexes
    • Lok HC, Suryo Rahmanto Y, Hawkins CL, Kalinowski DS, Morrow CS, Townsend AJ, Ponka P, Richardson DR. Nitric oxide storage and transport in cells are mediated by glutathione S-transferase P1-1 and multidrug resistance protein 1 via dinitrosyl iron complexes. J Biol Chem. 2012; 287:607-618.
    • (2012) J Biol Chem. , vol.287 , pp. 607-618
    • Lok, H.C.1    Suryo Rahmanto, Y.2    Hawkins, C.L.3    Kalinowski, D.S.4    Morrow, C.S.5    Townsend, A.J.6    Ponka, P.7    Richardson, D.R.8
  • 50
    • 33745479226 scopus 로고    scopus 로고
    • Nitrogen monoxide (NO)-mediated iron release from cells is linked to NO-induced glutathione efflux via multidrug resistance-associated protein 1
    • Watts RN, Hawkins C, Ponka P, Richardson DR. Nitrogen monoxide (NO)-mediated iron release from cells is linked to NO-induced glutathione efflux via multidrug resistance-associated protein 1. Proc Natl Acad Sci U S A. 2006; 103:7670-7675.
    • (2006) Proc Natl Acad Sci U S A. , vol.103 , pp. 7670-7675
    • Watts, R.N.1    Hawkins, C.2    Ponka, P.3    Richardson, D.R.4
  • 52
    • 0036133928 scopus 로고    scopus 로고
    • The serum ferritin concentration is a significant prognostic indicator of survival in primary lung cancer
    • Milman N, Pedersen LM. The serum ferritin concentration is a significant prognostic indicator of survival in primary lung cancer. Oncol Rep. 2002; 9:193-198.
    • (2002) Oncol Rep. , vol.9 , pp. 193-198
    • Milman, N.1    Pedersen, L.M.2
  • 53
    • 33846589444 scopus 로고    scopus 로고
    • Serum ferritin in colorectal cancer patients and its prognostic evaluation
    • Lorenzi M, Lorenzi B, Vernillo R. Serum ferritin in colorectal cancer patients and its prognostic evaluation. Int J Biol Markers. 2006; 21:235-241.
    • (2006) Int J Biol Markers. , vol.21 , pp. 235-241
    • Lorenzi, M.1    Lorenzi, B.2    Vernillo, R.3
  • 54
    • 0021994887 scopus 로고
    • Prognostic importance of serum ferritin in patients with Stages III and IV neuroblastoma: the Childrens Cancer Study Group experience
    • Hann HW, Evans AE, Siegel SE, Wong KY, Sather H, Dalton A, Hammond D, Seeger RC. Prognostic importance of serum ferritin in patients with Stages III and IV neuroblastoma: the Childrens Cancer Study Group experience. Cancer Res. 1985; 45:2843-2848.
    • (1985) Cancer Res. , vol.45 , pp. 2843-2848
    • Hann, H.W.1    Evans, A.E.2    Siegel, S.E.3    Wong, K.Y.4    Sather, H.5    Dalton, A.6    Hammond, D.7    Seeger, R.C.8
  • 55
    • 0033613854 scopus 로고    scopus 로고
    • Coordinated regulation of iron-controlling genes, H-ferritin and IRP2, by c-MYC
    • Wu KJ, Polack A, Dalla-Favera R. Coordinated regulation of iron-controlling genes, H-ferritin and IRP2, by c-MYC. Science. 1999; 283:676-679.
    • (1999) Science. , vol.283 , pp. 676-679
    • Wu, K.J.1    Polack, A.2    Dalla-Favera, R.3
  • 56
    • 0036565766 scopus 로고    scopus 로고
    • Ferritin expression modulates cell cycle dynamics and cell responsiveness to H-ras-induced growth via expansion of the labile iron pool
    • Kakhlon O, Gruenbaum Y, Cabantchik ZI. Ferritin expression modulates cell cycle dynamics and cell responsiveness to H-ras-induced growth via expansion of the labile iron pool. Biochem J. 2002; 363:431-436.
    • (2002) Biochem J. , vol.363 , pp. 431-436
    • Kakhlon, O.1    Gruenbaum, Y.2    Cabantchik, Z.I.3
  • 58
    • 0031001278 scopus 로고    scopus 로고
    • Iron-chelating therapy and the treatment of thalassemia
    • Olivieri NF, Brittenham GM. Iron-chelating therapy and the treatment of thalassemia. Blood. 1997; 89:739-761.
    • (1997) Blood. , vol.89 , pp. 739-761
    • Olivieri, N.F.1    Brittenham, G.M.2
  • 59
    • 0031869315 scopus 로고    scopus 로고
    • Development of iron chelators to treat iron overload disease and their use as experimental tools to probe intracellular iron metabolism
    • Richardson DR, Ponka P. Development of iron chelators to treat iron overload disease and their use as experimental tools to probe intracellular iron metabolism. Am J Hematol. 1998; 58:299-305.
    • (1998) Am J Hematol. , vol.58 , pp. 299-305
    • Richardson, D.R.1    Ponka, P.2
  • 62
    • 2942724321 scopus 로고    scopus 로고
    • Desferoxamine (DFO) - mediated iron chelation: rationale for a novel approach to therapy for brain cancer
    • Dayani PN, Bishop MC, Black K, Zeltzer PM. Desferoxamine (DFO) - mediated iron chelation: rationale for a novel approach to therapy for brain cancer. J Neuro Oncol. 2004; 67:367-377.
    • (2004) J Neuro Oncol. , vol.67 , pp. 367-377
    • Dayani, P.N.1    Bishop, M.C.2    Black, K.3    Zeltzer, P.M.4
  • 63
    • 0028840530 scopus 로고
    • Comparative efficacy and toxicity of desferrioxamine, deferiprone and other iron and aluminium chelating drugs
    • Kontoghiorghes GJ. Comparative efficacy and toxicity of desferrioxamine, deferiprone and other iron and aluminium chelating drugs. Toxicol Lett. 1995; 80:1-18.
    • (1995) Toxicol Lett. , vol.80 , pp. 1-18
    • Kontoghiorghes, G.J.1
  • 64
    • 0022871715 scopus 로고
    • Cytotoxic and DNA-inhibitory effects of iron chelators on human leukaemic cell lines
    • Kontoghiorghes GJ, Piga A, Hoffbrand AV. Cytotoxic and DNA-inhibitory effects of iron chelators on human leukaemic cell lines. Hematol Oncol. 1986; 4:195-204.
    • (1986) Hematol Oncol. , vol.4 , pp. 195-204
    • Kontoghiorghes, G.J.1    Piga, A.2    Hoffbrand, A.V.3
  • 66
    • 0023092624 scopus 로고
    • Antineuroblastoma activity of desferoxamine in human cell lines
    • Blatt J, Stitely S. Antineuroblastoma activity of desferoxamine in human cell lines. Cancer Res. 1987; 47:1749-1750.
    • (1987) Cancer Res. , vol.47 , pp. 1749-1750
    • Blatt, J.1    Stitely, S.2
  • 67
    • 0023689207 scopus 로고
    • Mechanism of antineuroblastoma activity of deferoxamine in vitro
    • Blatt J, Taylor SR, Stitely S. Mechanism of antineuroblastoma activity of deferoxamine in vitro. J Lab Clin Med. 1988; 112:433-436.
    • (1988) J Lab Clin Med. , vol.112 , pp. 433-436
    • Blatt, J.1    Taylor, S.R.2    Stitely, S.3
  • 69
    • 0029066979 scopus 로고
    • Deferoxamine followed by cyclophosphamide, etoposide, carboplatin, thiotepa, induction regimen in advanced neuroblastoma - preliminary results
    • Donfrancesco A, Debernardi B, Carli M, Mancini A, Nigro M, Desio L, Casale F, Bagnulo S, Helson L, Deb G. Deferoxamine followed by cyclophosphamide, etoposide, carboplatin, thiotepa, induction regimen in advanced neuroblastoma - preliminary results. Eur J Cancer. 1995; 31A:612-615.
    • (1995) Eur J Cancer , vol.31A , pp. 612-615
    • Donfrancesco, A.1    Debernardi, B.2    Carli, M.3    Mancini, A.4    Nigro, M.5    Desio, L.6    Casale, F.7    Bagnulo, S.8    Helson, L.9    Deb, G.10
  • 70
    • 0028585886 scopus 로고
    • Deferoxamine in children with recurrent neuroblastoma
    • Blatt J. Deferoxamine in children with recurrent neuroblastoma. Anticancer Res. 1994; 14:2109-2112.
    • (1994) Anticancer Res. , vol.14 , pp. 2109-2112
    • Blatt, J.1
  • 73
    • 0028158768 scopus 로고
    • The effect of the iron(III) chelator, desferrioxamine, on iron and transferrin uptake by the human malignant melanoma cell
    • Richardson D, Ponka P, Baker E. The effect of the iron(III) chelator, desferrioxamine, on iron and transferrin uptake by the human malignant melanoma cell. Cancer Res. 1994; 54:685-689.
    • (1994) Cancer Res. , vol.54 , pp. 685-689
    • Richardson, D.1    Ponka, P.2    Baker, E.3
  • 74
    • 70450219836 scopus 로고    scopus 로고
    • Deferasirox (Exjade) for the treatment of iron overload
    • Cappellini MD, Taher A. Deferasirox (Exjade) for the treatment of iron overload. Acta Haematol. 2009; 122:165-173.
    • (2009) Acta Haematol. , vol.122 , pp. 165-173
    • Cappellini, M.D.1    Taher, A.2
  • 78
    • 65349129409 scopus 로고    scopus 로고
    • The oral iron chelator deferasirox represses signaling through the mTOR in myeloid leukemia cells by enhancing expression of REDD1
    • Ohyashiki JH, Kobayashi C, Hamamura R, Okabe S, Tauchi T, Ohyashiki K. The oral iron chelator deferasirox represses signaling through the mTOR in myeloid leukemia cells by enhancing expression of REDD1. Cancer Sci. 2009; 100:970-977.
    • (2009) Cancer Sci. , vol.100 , pp. 970-977
    • Ohyashiki, J.H.1    Kobayashi, C.2    Hamamura, R.3    Okabe, S.4    Tauchi, T.5    Ohyashiki, K.6
  • 79
    • 34548675357 scopus 로고    scopus 로고
    • Antiproliferative and apoptotic effects in rat and human hepatoma cell cultures of the orally active iron chelator ICL670 compared to CP20: a possible relationship with polyamine metabolism
    • Lescoat G, Chantrel-Groussard K, Pasdeloup N, Nick H, Brissot P, Gaboriau F. Antiproliferative and apoptotic effects in rat and human hepatoma cell cultures of the orally active iron chelator ICL670 compared to CP20: a possible relationship with polyamine metabolism. Cell Prolif. 2007; 40:755-767.
    • (2007) Cell Prolif. , vol.40 , pp. 755-767
    • Lescoat, G.1    Chantrel-Groussard, K.2    Pasdeloup, N.3    Nick, H.4    Brissot, P.5    Gaboriau, F.6
  • 86
    • 0036269005 scopus 로고    scopus 로고
    • Iron chelators as therapeutic agents for the treatment of cancer
    • Richardson DR. Iron chelators as therapeutic agents for the treatment of cancer. Crit Rev Oncol Hematol. 2002; 42:267-281.
    • (2002) Crit Rev Oncol Hematol. , vol.42 , pp. 267-281
    • Richardson, D.R.1
  • 87
    • 0027934897 scopus 로고
    • Inhibitors of ribonucleotide reductase. Comparative effects of amino-and hydroxy-substituted pyridine-2-carboxaldehyde thiosemicarbazones
    • Cory JG, Cory AH, Rappa G, Lorico A, Liu MC, Lin TS, Sartorelli AC. Inhibitors of ribonucleotide reductase. Comparative effects of amino-and hydroxy-substituted pyridine-2-carboxaldehyde thiosemicarbazones. Biochem Pharmacol. 1994; 48:335-344.
    • (1994) Biochem Pharmacol. , vol.48 , pp. 335-344
    • Cory, J.G.1    Cory, A.H.2    Rappa, G.3    Lorico, A.4    Liu, M.C.5    Lin, T.S.6    Sartorelli, A.C.7
  • 89
    • 0034069521 scopus 로고    scopus 로고
    • Antiproliferative and apoptotic effects of O-Trensox, a new synthetic iron chelator, on differentiated human hepatoma cell lines
    • Rakba N, Loyer P, Gilot D, Delcros JG, Glaise D, Baret P, Pierre JL, Brissot P, Lescoat G. Antiproliferative and apoptotic effects of O-Trensox, a new synthetic iron chelator, on differentiated human hepatoma cell lines. Carcinogenesis. 2000; 21:943-951.
    • (2000) Carcinogenesis. , vol.21 , pp. 943-951
    • Rakba, N.1    Loyer, P.2    Gilot, D.3    Delcros, J.G.4    Glaise, D.5    Baret, P.6    Pierre, J.L.7    Brissot, P.8    Lescoat, G.9
  • 90
    • 0028891974 scopus 로고
    • The potential of iron chelators of the pyridoxal isonicotinoyl hydrazone class as effective antiproliferative agents
    • Richardson DR, Tran EH, Ponka P. The potential of iron chelators of the pyridoxal isonicotinoyl hydrazone class as effective antiproliferative agents. Blood. 1995; 86:4295-4306.
    • (1995) Blood. , vol.86 , pp. 4295-4306
    • Richardson, D.R.1    Tran, E.H.2    Ponka, P.3
  • 91
    • 0033566305 scopus 로고    scopus 로고
    • The potential of iron chelators of the pyridoxal isonicotinoyl hydrazone class as effective antiproliferative agents III: the effect of the ligands on molecular targets involved in proliferation
    • Darnell G, Richardson DR. The potential of iron chelators of the pyridoxal isonicotinoyl hydrazone class as effective antiproliferative agents III: the effect of the ligands on molecular targets involved in proliferation. Blood. 1999; 94:781-792.
    • (1999) Blood. , vol.94 , pp. 781-792
    • Darnell, G.1    Richardson, D.R.2
  • 92
    • 0037351292 scopus 로고    scopus 로고
    • Identification of the di-pyridyl ketone isonicotinoyl hydrazone (PKIH) analogues as potent iron chelators and anti-tumour agents
    • Becker EM, Lovejoy DB, Greer JM, Watts R, Richardson DR. Identification of the di-pyridyl ketone isonicotinoyl hydrazone (PKIH) analogues as potent iron chelators and anti-tumour agents. Br J Pharmacol. 2003; 138:819-830.
    • (2003) Br J Pharmacol. , vol.138 , pp. 819-830
    • Becker, E.M.1    Lovejoy, D.B.2    Greer, J.M.3    Watts, R.4    Richardson, D.R.5
  • 93
    • 0037100453 scopus 로고    scopus 로고
    • Novel "hybrid" iron chelators derived from aroylhydrazones and thiosemicarbazones demonstrate selective antiproliferative activity against tumor cells
    • Lovejoy DB, Richardson DR. Novel "hybrid" iron chelators derived from aroylhydrazones and thiosemicarbazones demonstrate selective antiproliferative activity against tumor cells. Blood. 2002; 100:666-676.
    • (2002) Blood. , vol.100 , pp. 666-676
    • Lovejoy, D.B.1    Richardson, D.R.2
  • 94
    • 0037237032 scopus 로고    scopus 로고
    • Examination of the antiproliferative activity of iron chelators: multiple cellular targets and the different mechanism of action of Triapine compared with desferrioxamine and the potent pyridoxal isonicotinoyl hydrazone analogue 311
    • Chaston TB, Lovejoy DB, Watts RN, Richardson DR. Examination of the antiproliferative activity of iron chelators: multiple cellular targets and the different mechanism of action of Triapine compared with desferrioxamine and the potent pyridoxal isonicotinoyl hydrazone analogue 311. Clin Cancer Res. 2003; 9:402-414.
    • (2003) Clin Cancer Res. , vol.9 , pp. 402-414
    • Chaston, T.B.1    Lovejoy, D.B.2    Watts, R.N.3    Richardson, D.R.4
  • 95
    • 4644288941 scopus 로고    scopus 로고
    • A phase I trial of 3-aminopyridine-2-carboxaldehyde thiosemicarbazone in combination with gemcitabine for patients with advanced cancer
    • Yen Y, Margolin K, Doroshow J, Fishman M, Johnson B, Clairmont C, Sullivan D, Sznol M. A phase I trial of 3-aminopyridine-2-carboxaldehyde thiosemicarbazone in combination with gemcitabine for patients with advanced cancer. Cancer Chemother Pharmacol. 2004; 54:331-342.
    • (2004) Cancer Chemother Pharmacol. , vol.54 , pp. 331-342
    • Yen, Y.1    Margolin, K.2    Doroshow, J.3    Fishman, M.4    Johnson, B.5    Clairmont, C.6    Sullivan, D.7    Sznol, M.8
  • 97
    • 34249888204 scopus 로고    scopus 로고
    • Phase I and pharmacokinetic study of Triapine (R), a potent ribonucleotide reductase inhibitor, in adults with advanced hematologic malignancies
    • Gojo I, Tidwell ML, Greer J, Takebe N, Seiter K, Pochron MF, Johnson B, Sznol M, Karp JE. Phase I and pharmacokinetic study of Triapine (R), a potent ribonucleotide reductase inhibitor, in adults with advanced hematologic malignancies. Leukemia Res. 2007; 31:1165-1173.
    • (2007) Leukemia Res. , vol.31 , pp. 1165-1173
    • Gojo, I.1    Tidwell, M.L.2    Greer, J.3    Takebe, N.4    Seiter, K.5    Pochron, M.F.6    Johnson, B.7    Sznol, M.8    Karp, J.E.9
  • 98
    • 2442698003 scopus 로고    scopus 로고
    • Phase I and pharmacokinetic study of the ribonucleotide reductase inhibitor, 3-aminopyridine-2-carboxaldehyde thiosemicarbazone, administered by 96-hour intravenous continuous infusion
    • Wadler S, Makower D, Clairmont C, Lambert P, Fehn K, Sznol M. Phase I and pharmacokinetic study of the ribonucleotide reductase inhibitor, 3-aminopyridine-2-carboxaldehyde thiosemicarbazone, administered by 96-hour intravenous continuous infusion. J Clin Oncol. 2004; 22:1553-1563.
    • (2004) J Clin Oncol. , vol.22 , pp. 1553-1563
    • Wadler, S.1    Makower, D.2    Clairmont, C.3    Lambert, P.4    Fehn, K.5    Sznol, M.6
  • 99
    • 34547687556 scopus 로고    scopus 로고
    • Phase II study of Triapine in patients with metastatic renal cell carcinoma: a trial of the National Cancer Institute of Canada Clinical Trials Group (NCIC IND.161)
    • Knox JJ, Hotte SJ, Kollmannsberger C, Winquist E, Fisher B, Eisenhauer EA. Phase II study of Triapine in patients with metastatic renal cell carcinoma: a trial of the National Cancer Institute of Canada Clinical Trials Group (NCIC IND.161). Invest New Drugs. 2007; 25:471-477.
    • (2007) Invest New Drugs. , vol.25 , pp. 471-477
    • Knox, J.J.1    Hotte, S.J.2    Kollmannsberger, C.3    Winquist, E.4    Fisher, B.5    Eisenhauer, E.A.6
  • 100
    • 40549137513 scopus 로고    scopus 로고
    • A multicenter phase II trial of 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP, Triapine) and gemcitabine in advanced non-small-cell lung cancer with pharmacokinetic evaluation using peripheral blood mononuclear cells
    • Ma B, Goh BC, Tan EH, Lam KC, Soo R, Leong SS, Wang LZ, Mo F, Chan AT, Zee B, Mok T. A multicenter phase II trial of 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP, Triapine) and gemcitabine in advanced non-small-cell lung cancer with pharmacokinetic evaluation using peripheral blood mononuclear cells. Invest New Drugs. 2008; 26:169-173.
    • (2008) Invest New Drugs. , vol.26 , pp. 169-173
    • Ma, B.1    Goh, B.C.2    Tan, E.H.3    Lam, K.C.4    Soo, R.5    Leong, S.S.6    Wang, L.Z.7    Mo, F.8    Chan, A.T.9    Zee, B.10    Mok, T.11
  • 101
    • 39549120057 scopus 로고    scopus 로고
    • A Phase II study of 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP) and gemcitabine in advanced pancreatic carcinoma. A trial of the Princess Margaret hospital Phase II consortium
    • Mackenzie MJ, Saltman D, Hirte H, Low J, Johnson C, Pond G, Moore MJ. A Phase II study of 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP) and gemcitabine in advanced pancreatic carcinoma. A trial of the Princess Margaret hospital Phase II consortium. Invest New Drugs. 2007; 25:553-558.
    • (2007) Invest New Drugs. , vol.25 , pp. 553-558
    • Mackenzie, M.J.1    Saltman, D.2    Hirte, H.3    Low, J.4    Johnson, C.5    Pond, G.6    Moore, M.J.7
  • 102
    • 74149093251 scopus 로고    scopus 로고
    • A phase II trial of Triapine (NSC# 663249) and gemcitabine as second line treatment of advanced non-small cell lung cancer: Eastern Cooperative Oncology Group Study 1503
    • Traynor AM, Lee JW, Bayer GK, Tate JM, Thomas SP, Mazurczak M, Graham DL, Kolesar JM, Schiller JH. A phase II trial of Triapine (NSC# 663249) and gemcitabine as second line treatment of advanced non-small cell lung cancer: Eastern Cooperative Oncology Group Study 1503. Invest New Drugs. 2010; 28:91-97.
    • (2010) Invest New Drugs. , vol.28 , pp. 91-97
    • Traynor, A.M.1    Lee, J.W.2    Bayer, G.K.3    Tate, J.M.4    Thomas, S.P.5    Mazurczak, M.6    Graham, D.L.7    Kolesar, J.M.8    Schiller, J.H.9
  • 103
    • 0030894798 scopus 로고    scopus 로고
    • The potential of iron chelators of the pyridoxal isonicotinoyl hydrazone class as effective antiproliferative agents II: the mechanism of action of ligands derived from salicylaldehyde benzoyl hydrazone and 2-hydroxy-1-naphthylaldehyde benzoyl hydrazone
    • Richardson DR, Milnes K. The potential of iron chelators of the pyridoxal isonicotinoyl hydrazone class as effective antiproliferative agents II: the mechanism of action of ligands derived from salicylaldehyde benzoyl hydrazone and 2-hydroxy-1-naphthylaldehyde benzoyl hydrazone. Blood. 1997; 89:3025-3038.
    • (1997) Blood. , vol.89 , pp. 3025-3038
    • Richardson, D.R.1    Milnes, K.2
  • 104
    • 0035437183 scopus 로고    scopus 로고
    • The potential of iron chelators of the pyridoxal isonicotinoyl hydrazone class as effective antiproliferative agents, IV: the mechanisms involved in inhibiting cell-cycle progression
    • Gao J, Richardson DR. The potential of iron chelators of the pyridoxal isonicotinoyl hydrazone class as effective antiproliferative agents, IV: the mechanisms involved in inhibiting cell-cycle progression. Blood. 2001; 98:842-850.
    • (2001) Blood. , vol.98 , pp. 842-850
    • Gao, J.1    Richardson, D.R.2
  • 105
    • 4444248453 scopus 로고    scopus 로고
    • Novel di-2-pyridyl-derived iron chelators with marked and selective antitumor activity: in vitro and in vivo assessment
    • Yuan J, Lovejoy DB, Richardson DR. Novel di-2-pyridyl-derived iron chelators with marked and selective antitumor activity: in vitro and in vivo assessment. Blood. 2004; 104:1450-1458.
    • (2004) Blood. , vol.104 , pp. 1450-1458
    • Yuan, J.1    Lovejoy, D.B.2    Richardson, D.R.3
  • 106
    • 33749515083 scopus 로고    scopus 로고
    • A class of iron chelators with a wide spectrum of potent antitumor activity that overcomes resistance to chemotherapeutics
    • Whitnall M, Howard J, Ponka P, Richardson DR. A class of iron chelators with a wide spectrum of potent antitumor activity that overcomes resistance to chemotherapeutics. Proc Natl Acad Sci U S A. 2006; 103:14901-14906.
    • (2006) Proc Natl Acad Sci U S A. , vol.103 , pp. 14901-14906
    • Whitnall, M.1    Howard, J.2    Ponka, P.3    Richardson, D.R.4
  • 109
    • 80053198672 scopus 로고    scopus 로고
    • Novel thiosemicarbazone iron chelators induce up-regulation and phosphorylation of the metastasis suppressor N-myc down-stream regulated gene 1: a new strategy for the treatment of pancreatic cancer
    • Kovacevic Z, Chikhani S, Lovejoy DB, Richardson DR. Novel thiosemicarbazone iron chelators induce up-regulation and phosphorylation of the metastasis suppressor N-myc down-stream regulated gene 1: a new strategy for the treatment of pancreatic cancer. Mol Pharmacol. 2011; 80:598-609.
    • (2011) Mol Pharmacol. , vol.80 , pp. 598-609
    • Kovacevic, Z.1    Chikhani, S.2    Lovejoy, D.B.3    Richardson, D.R.4
  • 110
    • 80052221642 scopus 로고    scopus 로고
    • Antitumor activity of metal-chelating compound Dp44mT is mediated by formation of a redox-active copper complex that accumulates in lysosomes
    • Lovejoy DB, Jansson PJ, Brunk UT, Wong J, Ponka P, Richardson DR. Antitumor activity of metal-chelating compound Dp44mT is mediated by formation of a redox-active copper complex that accumulates in lysosomes. Cancer Res. 2011; 71:5871-5880.
    • (2011) Cancer Res. , vol.71 , pp. 5871-5880
    • Lovejoy, D.B.1    Jansson, P.J.2    Brunk, U.T.3    Wong, J.4    Ponka, P.5    Richardson, D.R.6
  • 111
  • 112
    • 84926617610 scopus 로고    scopus 로고
    • Di-2-pyridylketone 4, 4-Dimethyl-3-thiosemicarbazone (Dp44mT) overcomes multidrug-resistance by a novel mechanism involving the hijacking of lysosomal P-Glycoprotein (Pgp)
    • Jansson PJ, Yamagishi T, Arvind A, Seebacher N, Gutierrez E, Stacy A, Maleki S, Sharp D, Sahni S, Richardson DR. Di-2-pyridylketone 4, 4-Dimethyl-3-thiosemicarbazone (Dp44mT) overcomes multidrug-resistance by a novel mechanism involving the hijacking of lysosomal P-Glycoprotein (Pgp). J Biol Chem. 2015; 290:9588-9603.
    • (2015) J Biol Chem. , vol.290 , pp. 9588-9603
    • Jansson, P.J.1    Yamagishi, T.2    Arvind, A.3    Seebacher, N.4    Gutierrez, E.5    Stacy, A.6    Maleki, S.7    Sharp, D.8    Sahni, S.9    Richardson, D.R.10
  • 113
    • 84866887477 scopus 로고    scopus 로고
    • Novel second-generation di-2-pyridylketone thiosemicarbazones show synergism with standard chemotherapeutics and demonstrate potent activity against lung cancer xenografts after oral and intravenous administration in vivo
    • Lovejoy DB, Sharp DM, Seebacher N, Obeidy P, Prichard T, Stefani C, Basha MT, Sharpe PC, Jansson PJ, Kalinowski DS, Bernhardt PV, Richardson DR. Novel second-generation di-2-pyridylketone thiosemicarbazones show synergism with standard chemotherapeutics and demonstrate potent activity against lung cancer xenografts after oral and intravenous administration in vivo. J Med Chem. 2012; 55:7230-7244.
    • (2012) J Med Chem. , vol.55 , pp. 7230-7244
    • Lovejoy, D.B.1    Sharp, D.M.2    Seebacher, N.3    Obeidy, P.4    Prichard, T.5    Stefani, C.6    Basha, M.T.7    Sharpe, P.C.8    Jansson, P.J.9    Kalinowski, D.S.10    Bernhardt, P.V.11    Richardson, D.R.12
  • 114
    • 34548286520 scopus 로고    scopus 로고
    • Tuning cell cycle regulation with an iron key
    • Yu Y, Kovacevic Z, Richardson DR. Tuning cell cycle regulation with an iron key. Cell Cycle. 2007; 6:1982-1994.
    • (2007) Cell Cycle. , vol.6 , pp. 1982-1994
    • Yu, Y.1    Kovacevic, Z.2    Richardson, D.R.3
  • 118
    • 0027521831 scopus 로고
    • Role of ribonucleotide reductase in inhibition of mammalian cell growth by potent iron chelators
    • Nyholm S, Mann GJ, Johansson AG, Bergeron RJ, Graslund A, Thelander L. Role of ribonucleotide reductase in inhibition of mammalian cell growth by potent iron chelators. J Biol Chem. 1993; 268:26200-26205.
    • (1993) J Biol Chem. , vol.268 , pp. 26200-26205
    • Nyholm, S.1    Mann, G.J.2    Johansson, A.G.3    Bergeron, R.J.4    Graslund, A.5    Thelander, L.6
  • 119
    • 0036684318 scopus 로고    scopus 로고
    • Evolving role of ribonucleoside reductase inhibitors in hematologic malignancies
    • Tsimberidou A-M, Alvarado Y, Giles FJ. Evolving role of ribonucleoside reductase inhibitors in hematologic malignancies. Expert Rev Anticancer Ther. 2002; 2:437-448.
    • (2002) Expert Rev Anticancer Ther. , vol.2 , pp. 437-448
    • Tsimberidou, A.-M.1    Alvarado, Y.2    Giles, F.J.3
  • 120
    • 84873737776 scopus 로고    scopus 로고
    • Dp44mT targets the AKT, TGF-beta and ERK pathways via the metastasis suppressor NDRG1 in normal prostate epithelial cells and prostate cancer cells
    • Dixon KM, Lui GY, Kovacevic Z, Zhang D, Yao M, Chen Z, Dong Q, Assinder SJ, Richardson DR. Dp44mT targets the AKT, TGF-beta and ERK pathways via the metastasis suppressor NDRG1 in normal prostate epithelial cells and prostate cancer cells. Br J Cancer. 2013; 108:409-419.
    • (2013) Br J Cancer. , vol.108 , pp. 409-419
    • Dixon, K.M.1    Lui, G.Y.2    Kovacevic, Z.3    Zhang, D.4    Yao, M.5    Chen, Z.6    Dong, Q.7    Assinder, S.J.8    Richardson, D.R.9
  • 121
    • 84925283220 scopus 로고    scopus 로고
    • Novel thiosemicarbazones regulate the signal transducer and activator of transcription 3 (STAT3) pathway: inhibition of constitutive and interleukin 6-induced activation by iron depletion
    • Lui GYL, Kovacevic Z, S VM, Kalinowski DS, Merlot AM, Sahni S, Richardson DR. Novel thiosemicarbazones regulate the signal transducer and activator of transcription 3 (STAT3) pathway: inhibition of constitutive and interleukin 6-induced activation by iron depletion. Mol Pharmacol. 2015; 87:543-560.
    • (2015) Mol Pharmacol. , vol.87 , pp. 543-560
    • Lui, G.Y.L.1    Kovacevic, Z.2    Vm, S.3    Kalinowski, D.S.4    Merlot, A.M.5    Sahni, S.6    Richardson, D.R.7
  • 122
    • 0030601987 scopus 로고    scopus 로고
    • Iron deprivation inhibits cyclin-dependent kinase activity and decreases cyclin D CDK4 protein levels in asynchronous MDA-MB-453 human breast cancer cells
    • Kulp KS, Green SL, Vulliet PR. Iron deprivation inhibits cyclin-dependent kinase activity and decreases cyclin D CDK4 protein levels in asynchronous MDA-MB-453 human breast cancer cells. Exp Cell Res. 1996; 229:60-68.
    • (1996) Exp Cell Res. , vol.229 , pp. 60-68
    • Kulp, K.S.1    Green, S.L.2    Vulliet, P.R.3
  • 124
    • 0028927179 scopus 로고
    • p53, cell-cycle control and apoptosis - implications for cancer
    • Kastan MB, Canman CE, Leonard CJ. p53, cell-cycle control and apoptosis - implications for cancer. Cancer Metast Rev. 1995; 14:3-15.
    • (1995) Cancer Metast Rev. , vol.14 , pp. 3-15
    • Kastan, M.B.1    Canman, C.E.2    Leonard, C.J.3
  • 125
    • 0142154312 scopus 로고    scopus 로고
    • The effect of potent iron chelators on the regulation of p53: examination of the expression, localization and DNA-binding activity of p53 and the transactivation of WAF1
    • Liang SX, Richardson DR. The effect of potent iron chelators on the regulation of p53: examination of the expression, localization and DNA-binding activity of p53 and the transactivation of WAF1. Carcinogenesis. 2003; 24:1601-1614.
    • (2003) Carcinogenesis. , vol.24 , pp. 1601-1614
    • Liang, S.X.1    Richardson, D.R.2
  • 128
    • 0347628818 scopus 로고    scopus 로고
    • p21(Waf1/Cip1) as a therapeutic target in breast and other cancers
    • Weiss RH. p21(Waf1/Cip1) as a therapeutic target in breast and other cancers. Cancer Cell. 2003; 4:425-429.
    • (2003) Cancer Cell. , vol.4 , pp. 425-429
    • Weiss, R.H.1
  • 129
    • 33750903389 scopus 로고    scopus 로고
    • Requirement for pre-existing of p21 to prevent doxorubicin-induced apoptosis through inhibition of caspase-3 activation
    • Tang JJ, Shen C, Lu YJ. Requirement for pre-existing of p21 to prevent doxorubicin-induced apoptosis through inhibition of caspase-3 activation. Mol Cell Biochem. 2006; 291:139-144.
    • (2006) Mol Cell Biochem. , vol.291 , pp. 139-144
    • Tang, J.J.1    Shen, C.2    Lu, Y.J.3
  • 130
    • 34547137681 scopus 로고    scopus 로고
    • Iron chelation and regulation of the cell cycle: 2 mechanisms of posttranscriptional regulation of the universal cyclin-dependent kinase inhibitor p21(CIP1/WAF1) by iron depletion
    • Fu D, Richardson DR. Iron chelation and regulation of the cell cycle: 2 mechanisms of posttranscriptional regulation of the universal cyclin-dependent kinase inhibitor p21(CIP1/WAF1) by iron depletion. Blood. 2007; 110:752-761.
    • (2007) Blood. , vol.110 , pp. 752-761
    • Fu, D.1    Richardson, D.R.2
  • 131
    • 0141844609 scopus 로고    scopus 로고
    • The cyclin-dependent kinase inhibitor p21(CIP/WAF) is a positive regulator of insulin-like growth factor I-induced cell proliferation in MCF-7 human breast cancer cells
    • Dupont JL, Karas M, LeRoith D. The cyclin-dependent kinase inhibitor p21(CIP/WAF) is a positive regulator of insulin-like growth factor I-induced cell proliferation in MCF-7 human breast cancer cells. J Biol Chem. 2003; 278:37256-37264.
    • (2003) J Biol Chem. , vol.278 , pp. 37256-37264
    • Dupont, J.L.1    Karas, M.2    LeRoith, D.3
  • 132
    • 84928927872 scopus 로고    scopus 로고
    • Mammalian MYC Proteins and Cancer
    • Tansey WP. Mammalian MYC Proteins and Cancer. New J Sci. 2014; 2014:757534.
    • (2014) New J Sci. , vol.2014 , pp. 757534
    • Tansey, W.P.1
  • 134
    • 84859171807 scopus 로고    scopus 로고
    • MYC on the path to cancer
    • Dang CV. MYC on the path to cancer. Cell. 2012; 149:22-35.
    • (2012) Cell. , vol.149 , pp. 22-35
    • Dang, C.V.1
  • 136
    • 0032996623 scopus 로고    scopus 로고
    • c-Myc regulates cyclin D-Cdk4 and -Cdk6 activity but affects cell cycle progression at multiple independent points
    • Mateyak MK, Obaya AJ, Sedivy JM. c-Myc regulates cyclin D-Cdk4 and -Cdk6 activity but affects cell cycle progression at multiple independent points. Mol Cell Biol. 1999; 19:4672-4683.
    • (1999) Mol Cell Biol. , vol.19 , pp. 4672-4683
    • Mateyak, M.K.1    Obaya, A.J.2    Sedivy, J.M.3
  • 138
    • 0031973694 scopus 로고    scopus 로고
    • Decreased expression of c-myc oncoprotein by peripheral blood mononuclear cells in thalassaemia patients receiving desferrioxamine
    • Kyriakou D, Eliopoulos AG, Papadakis A, Alexandrakis M, Eliopoulos GD. Decreased expression of c-myc oncoprotein by peripheral blood mononuclear cells in thalassaemia patients receiving desferrioxamine. Eur J Haematol. 1998; 60:21-27.
    • (1998) Eur J Haematol. , vol.60 , pp. 21-27
    • Kyriakou, D.1    Eliopoulos, A.G.2    Papadakis, A.3    Alexandrakis, M.4    Eliopoulos, G.D.5
  • 139
    • 33845358660 scopus 로고    scopus 로고
    • The metastasis suppressor, Ndrg-1: a new ally in the fight against cancer
    • Kovacevic Z, Richardson DR. The metastasis suppressor, Ndrg-1: a new ally in the fight against cancer. Carcinogenesis. 2006; 27:2355-2366.
    • (2006) Carcinogenesis. , vol.27 , pp. 2355-2366
    • Kovacevic, Z.1    Richardson, D.R.2
  • 140
    • 38849111785 scopus 로고    scopus 로고
    • NDRG1, a growth and cancer related gene: regulation of gene expression and function in normal and disease states
    • Ellen TP, Ke Q, Zhang P, Costa M. NDRG1, a growth and cancer related gene: regulation of gene expression and function in normal and disease states. Carcinogenesis. 2008; 29:2-8.
    • (2008) Carcinogenesis. , vol.29 , pp. 2-8
    • Ellen, T.P.1    Ke, Q.2    Zhang, P.3    Costa, M.4
  • 144
    • 0033972140 scopus 로고    scopus 로고
    • Drg-1 as a differentiation-related, putative metastatic suppressor gene in human colon cancer
    • Guang RJ, Ford JL, Fu YN, Li YZ, Shaw LM, Pardee AB. Drg-1 as a differentiation-related, putative metastatic suppressor gene in human colon cancer. Cancer Res. 2000; 60:749-755.
    • (2000) Cancer Res. , vol.60 , pp. 749-755
    • Guang, R.J.1    Ford, J.L.2    Fu, Y.N.3    Li, Y.Z.4    Shaw, L.M.5    Pardee, A.B.6
  • 145
    • 33745700194 scopus 로고    scopus 로고
    • Tumor growth suppression in pancreatic cancer by a putative metastasis suppressor gene Cap43/NDRG1/Drg-1 through modulation of angiogenesis
    • Maruyama Y, Ono M, Kawahara A, Yokoyama T, Basaki Y, Kage M, Aoyagi S, Kinoshita H, Kuwano M. Tumor growth suppression in pancreatic cancer by a putative metastasis suppressor gene Cap43/NDRG1/Drg-1 through modulation of angiogenesis. Cancer Res. 2006; 66:6233-6242.
    • (2006) Cancer Res. , vol.66 , pp. 6233-6242
    • Maruyama, Y.1    Ono, M.2    Kawahara, A.3    Yokoyama, T.4    Basaki, Y.5    Kage, M.6    Aoyagi, S.7    Kinoshita, H.8    Kuwano, M.9
  • 146
    • 7244239197 scopus 로고    scopus 로고
    • Iron chelators with high antiproliferative activity up-regulate the expression of a growth inhibitory and metastasis suppressor gene: a link between iron metabolism and proliferation
    • Le NTV, Richardson DR. Iron chelators with high antiproliferative activity up-regulate the expression of a growth inhibitory and metastasis suppressor gene: a link between iron metabolism and proliferation. Blood. 2004; 104:2967-2975.
    • (2004) Blood. , vol.104 , pp. 2967-2975
    • Le, N.T.V.1    Richardson, D.R.2
  • 147
    • 50849085751 scopus 로고    scopus 로고
    • The iron-regulated metastasis suppressor, Ndrg-1: identification of novel molecular targets
    • Kovacevic Z, Fu D, Richardson DR. The iron-regulated metastasis suppressor, Ndrg-1: identification of novel molecular targets. Biochim Biophys Acta. 2008; 1783:1981-1992.
    • (2008) Biochim Biophys Acta. , vol.1783 , pp. 1981-1992
    • Kovacevic, Z.1    Fu, D.2    Richardson, D.R.3
  • 148
    • 84874303394 scopus 로고    scopus 로고
    • N-myc downstream regulated 1 (NDRG1) is regulated by eukaryotic initiation factor 3a (eIF3a) during cellular stress caused by iron depletion
    • Lane DJ, Saletta F, Suryo Rahmanto Y, Kovacevic Z, Richardson DR. N-myc downstream regulated 1 (NDRG1) is regulated by eukaryotic initiation factor 3a (eIF3a) during cellular stress caused by iron depletion. PLoS One. 2013; 8:e57273.
    • (2013) PLoS One. , vol.8 , pp. e57273
    • Lane, D.J.1    Saletta, F.2    Suryo Rahmanto, Y.3    Kovacevic, Z.4    Richardson, D.R.5
  • 150
    • 77952745809 scopus 로고    scopus 로고
    • Identification of sites subjected to serine/threonine phosphorylation by SGK1 affecting N-myc downstream-regulated gene 1 (NDRG1)/Cap43-dependent suppression of angiogenic CXC chemokine expression in human pancreatic cancer cells
    • Murakami Y, Hosoi F, Izumi H, Maruyama Y, Ureshino H, Watari K, Kohno K, Kuwano M, Ono M. Identification of sites subjected to serine/threonine phosphorylation by SGK1 affecting N-myc downstream-regulated gene 1 (NDRG1)/Cap43-dependent suppression of angiogenic CXC chemokine expression in human pancreatic cancer cells. Biochem Biophys Res Commun. 2010; 396:376-381.
    • (2010) Biochem Biophys Res Commun. , vol.396 , pp. 376-381
    • Murakami, Y.1    Hosoi, F.2    Izumi, H.3    Maruyama, Y.4    Ureshino, H.5    Watari, K.6    Kohno, K.7    Kuwano, M.8    Ono, M.9
  • 151
    • 79960833879 scopus 로고    scopus 로고
    • Phosphorylation of NDRG1 is temporally and spatially controlled during the cell cycle
    • McCaig C, Potter L, Abramczyk O, Murray JT. Phosphorylation of NDRG1 is temporally and spatially controlled during the cell cycle. Biochem Biophys Res Commun. 2011; 411:227-234.
    • (2011) Biochem Biophys Res Commun. , vol.411 , pp. 227-234
    • McCaig, C.1    Potter, L.2    Abramczyk, O.3    Murray, J.T.4
  • 152
    • 84879671984 scopus 로고    scopus 로고
    • Proteolytic cleavage and truncation of NDRG1 in human prostate cancer cells, but not normal prostate epithelial cells
    • art:e00042
    • Ghalayini MK, Dong Q, Richardson DR, Assinder SJ. Proteolytic cleavage and truncation of NDRG1 in human prostate cancer cells, but not normal prostate epithelial cells. Biosci Rep. 2013; 33. art:e00042 .doi: 00010.01042/BSR20130042.
    • (2013) Biosci Rep , vol.33
    • Ghalayini, M.K.1    Dong, Q.2    Richardson, D.R.3    Assinder, S.J.4
  • 155
    • 79955770381 scopus 로고    scopus 로고
    • The metastasis suppressor, N-myc downstream regulated gene 1 (NDRG1), upregulates p21 via p53-independent mechanisms
    • Kovacevic Z, Sivagurunathan S, Mangs H, Chikhani S, Zhang D, Richardson DR. The metastasis suppressor, N-myc downstream regulated gene 1 (NDRG1), upregulates p21 via p53-independent mechanisms. Carcinogenesis. 2011; 32:732-740.
    • (2011) Carcinogenesis. , vol.32 , pp. 732-740
    • Kovacevic, Z.1    Sivagurunathan, S.2    Mangs, H.3    Chikhani, S.4    Zhang, D.5    Richardson, D.R.6
  • 156
    • 84861212961 scopus 로고    scopus 로고
    • The iron chelators Dp44mT and DFO inhibit TGF-beta-induced epithelial-mesenchymal transition via up-regulation of N-Myc downstream-regulated gene 1 (NDRG1)
    • Chen Z, Zhang D, Yue F, Zheng M, Kovacevic Z, Richardson DR. The iron chelators Dp44mT and DFO inhibit TGF-beta-induced epithelial-mesenchymal transition via up-regulation of N-Myc downstream-regulated gene 1 (NDRG1). J Biol Chem. 2012; 287:17016-17028.
    • (2012) J Biol Chem. , vol.287 , pp. 17016-17028
    • Chen, Z.1    Zhang, D.2    Yue, F.3    Zheng, M.4    Kovacevic, Z.5    Richardson, D.R.6
  • 157
    • 84872735807 scopus 로고    scopus 로고
    • Targeting the metastasis suppressor, NDRG1, using novel iron chelators: regulation of stress fiber-mediated tumor cell migration via modulation of the ROCK1/pMLC2 signaling pathway
    • Sun J, Zhang D, Zheng Y, Zhao Q, Zheng M, Kovacevic Z, Richardson DR. Targeting the metastasis suppressor, NDRG1, using novel iron chelators: regulation of stress fiber-mediated tumor cell migration via modulation of the ROCK1/pMLC2 signaling pathway. Mol Pharmacol. 2013; 83:454-469.
    • (2013) Mol Pharmacol. , vol.83 , pp. 454-469
    • Sun, J.1    Zhang, D.2    Zheng, Y.3    Zhao, Q.4    Zheng, M.5    Kovacevic, Z.6    Richardson, D.R.7
  • 158
    • 54949109808 scopus 로고    scopus 로고
    • PI3K/Akt: getting it right matters
    • Franke TF. PI3K/Akt: getting it right matters. Oncogene. 2008; 27:6473-6488.
    • (2008) Oncogene. , vol.27 , pp. 6473-6488
    • Franke, T.F.1
  • 160
    • 34250788809 scopus 로고    scopus 로고
    • AKT/PKB signaling: navigating downstream
    • Manning BD, Cantley LC. AKT/PKB signaling: navigating downstream. Cell. 2007; 129:1261-1274.
    • (2007) Cell. , vol.129 , pp. 1261-1274
    • Manning, B.D.1    Cantley, L.C.2
  • 161
    • 66149141021 scopus 로고    scopus 로고
    • PTEN and the PI3-kinase pathway in cancer
    • Chalhoub N, Baker SJ. PTEN and the PI3-kinase pathway in cancer. Annu Rev Pathol. 2009; 4:127-150.
    • (2009) Annu Rev Pathol. , vol.4 , pp. 127-150
    • Chalhoub, N.1    Baker, S.J.2
  • 163
    • 0033551070 scopus 로고    scopus 로고
    • New insights into tumor suppression: PTEN suppresses tumor formation by restraining the phosphoinositide 3-kinase/AKT pathway
    • Cantley LC, Neel BG. New insights into tumor suppression: PTEN suppresses tumor formation by restraining the phosphoinositide 3-kinase/AKT pathway. Proc Natl Acad Sci US A. 1999; 96:4240-4245.
    • (1999) Proc Natl Acad Sci US A. , vol.96 , pp. 4240-4245
    • Cantley, L.C.1    Neel, B.G.2
  • 165
    • 0030702123 scopus 로고    scopus 로고
    • Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery
    • Datta SR, Dudek H, Tao X, Masters S, Fu H, Gotoh Y, Greenberg ME. Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell. 1997; 91:231-241.
    • (1997) Cell. , vol.91 , pp. 231-241
    • Datta, S.R.1    Dudek, H.2    Tao, X.3    Masters, S.4    Fu, H.5    Gotoh, Y.6    Greenberg, M.E.7
  • 166
    • 35148861084 scopus 로고    scopus 로고
    • beta-catenin regulates multiple steps of RNA metabolism as revealed by the RNA aptamer in colon cancer cells
    • Lee HK, Kwak HY, Hur J, Kim IA, Yang JS, Park MW, Yu J, Jeong S. beta-catenin regulates multiple steps of RNA metabolism as revealed by the RNA aptamer in colon cancer cells. Cancer Res. 2007; 67:9315-9321.
    • (2007) Cancer Res. , vol.67 , pp. 9315-9321
    • Lee, H.K.1    Kwak, H.Y.2    Hur, J.3    Kim, I.A.4    Yang, J.S.5    Park, M.W.6    Yu, J.7    Jeong, S.8
  • 167
    • 0032533225 scopus 로고    scopus 로고
    • Glycogen synthase kinase-3β regulates cyclin D1 proteolysis and subcellular localization
    • Diehl JA, Cheng M, Roussel MF, Sherr CJ. Glycogen synthase kinase-3β regulates cyclin D1 proteolysis and subcellular localization. Genes Dev. 1998; 12:3499-3511.
    • (1998) Genes Dev. , vol.12 , pp. 3499-3511
    • Diehl, J.A.1    Cheng, M.2    Roussel, M.F.3    Sherr, C.J.4
  • 168
    • 0032484019 scopus 로고    scopus 로고
    • CREB is a regulatory target for the protein kinase Akt/PKB
    • Du KY, Montminy M. CREB is a regulatory target for the protein kinase Akt/PKB. J Biol Chem. 1998; 273:32377-32379.
    • (1998) J Biol Chem. , vol.273 , pp. 32377-32379
    • Du, K.Y.1    Montminy, M.2
  • 169
    • 0031433335 scopus 로고    scopus 로고
    • Phosphatidylinositol 3-kinase couples the interleukin-2 receptor to the cell cycle regulator E2F
    • Brennan P, Babbage JW, Burgering BMT, Groner B, Reif K, Cantrell DA. Phosphatidylinositol 3-kinase couples the interleukin-2 receptor to the cell cycle regulator E2F. Immunity. 1997; 7:679-689.
    • (1997) Immunity. , vol.7 , pp. 679-689
    • Brennan, P.1    Babbage, J.W.2    Burgering, B.M.T.3    Groner, B.4    Reif, K.5    Cantrell, D.A.6
  • 172
    • 0034462121 scopus 로고    scopus 로고
    • Forkhead transcription factors are critical effectors of cell death and cell cycle arrest downstream of PTEN
    • Nakamura N, Ramaswamy S, Vazquez F, Signoretti S, Loda M, Sellers WR. Forkhead transcription factors are critical effectors of cell death and cell cycle arrest downstream of PTEN. Mol Cell Biol. 2000; 20:8969-8982.
    • (2000) Mol Cell Biol. , vol.20 , pp. 8969-8982
    • Nakamura, N.1    Ramaswamy, S.2    Vazquez, F.3    Signoretti, S.4    Loda, M.5    Sellers, W.R.6
  • 173
    • 0034234924 scopus 로고    scopus 로고
    • A direct linkage between the phosphoinositide 3-kinase-AKT signaling pathway and the mammalian target of rapamycin in mitogen-stimulated and transformed cells
    • Sekulic A, Hudson CC, Homme JL, Yin P, Otterness DM, Karnitz LM, Abraham RT. A direct linkage between the phosphoinositide 3-kinase-AKT signaling pathway and the mammalian target of rapamycin in mitogen-stimulated and transformed cells. Cancer Res. 2000; 60:3504-3513.
    • (2000) Cancer Res. , vol.60 , pp. 3504-3513
    • Sekulic, A.1    Hudson, C.C.2    Homme, J.L.3    Yin, P.4    Otterness, D.M.5    Karnitz, L.M.6    Abraham, R.T.7
  • 174
    • 0036713778 scopus 로고    scopus 로고
    • TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling
    • Inoki K, Li Y, Zhu T, Wu J, Guan KL. TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nat Cell Biol. 2002; 4:648-657.
    • (2002) Nat Cell Biol. , vol.4 , pp. 648-657
    • Inoki, K.1    Li, Y.2    Zhu, T.3    Wu, J.4    Guan, K.L.5
  • 175
    • 0036342294 scopus 로고    scopus 로고
    • Identification of the tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the phosphoinositide 3-kinase/Akt pathway
    • Manning BD, Tee AR, Logsdon MN, Blenis J, Cantley LC. Identification of the tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the phosphoinositide 3-kinase/Akt pathway. Mol Cell. 2002; 10:151-162.
    • (2002) Mol Cell. , vol.10 , pp. 151-162
    • Manning, B.D.1    Tee, A.R.2    Logsdon, M.N.3    Blenis, J.4    Cantley, L.C.5
  • 176
    • 59749091850 scopus 로고    scopus 로고
    • A complex interplay between Akt, TSC2 and the two mTOR complexes
    • Huang J, Manning BD. A complex interplay between Akt, TSC2 and the two mTOR complexes. Biochem Soc Trans. 2009; 37:217-222.
    • (2009) Biochem Soc Trans. , vol.37 , pp. 217-222
    • Huang, J.1    Manning, B.D.2
  • 177
    • 4043171462 scopus 로고    scopus 로고
    • Upstream and downstream of mTOR
    • Hay N, Sonenberg N. Upstream and downstream of mTOR. Genes Dev. 2004; 18:1926-1945.
    • (2004) Genes Dev. , vol.18 , pp. 1926-1945
    • Hay, N.1    Sonenberg, N.2
  • 178
    • 0032514376 scopus 로고    scopus 로고
    • Translational induction of the c-myc oncogene via activation of the FRAP/TOR signalling pathway
    • West MJ, Stoneley M, Willis AE. Translational induction of the c-myc oncogene via activation of the FRAP/TOR signalling pathway. Oncogene. 1998; 17:769-780.
    • (1998) Oncogene. , vol.17 , pp. 769-780
    • West, M.J.1    Stoneley, M.2    Willis, A.E.3
  • 179
    • 18144399578 scopus 로고    scopus 로고
    • mTOR-targeted therapy of cancer with rapamycin derivatives
    • Vignot S, Faivre S, Aguirre D, Raymond E. mTOR-targeted therapy of cancer with rapamycin derivatives. Ann Oncol. 2005; 16:525-537.
    • (2005) Ann Oncol. , vol.16 , pp. 525-537
    • Vignot, S.1    Faivre, S.2    Aguirre, D.3    Raymond, E.4
  • 180
    • 67749122122 scopus 로고    scopus 로고
    • Targeting PI3K signalling in cancer: opportunities, challenges and limitations
    • Engelman JA. Targeting PI3K signalling in cancer: opportunities, challenges and limitations. Nat Rev Cancer. 2009; 9:550-562.
    • (2009) Nat Rev Cancer. , vol.9 , pp. 550-562
    • Engelman, J.A.1
  • 184
    • 33646706052 scopus 로고    scopus 로고
    • Oncogenic PI3K and its role in cancer
    • Samuels Y, Ericson K. Oncogenic PI3K and its role in cancer. Curr Opin Oncol. 2006; 18:77-82.
    • (2006) Curr Opin Oncol. , vol.18 , pp. 77-82
    • Samuels, Y.1    Ericson, K.2
  • 185
    • 39649113108 scopus 로고    scopus 로고
    • Interleukin-6 activates PI3K/Akt pathway and regulates cyclin A1 to promote prostate cancer cell survival
    • Wegiel B, Bjartell A, Culig Z, Persson JL. Interleukin-6 activates PI3K/Akt pathway and regulates cyclin A1 to promote prostate cancer cell survival. Int J Cancer. 2008; 122:1521-1529.
    • (2008) Int J Cancer. , vol.122 , pp. 1521-1529
    • Wegiel, B.1    Bjartell, A.2    Culig, Z.3    Persson, J.L.4
  • 187
    • 84873882079 scopus 로고    scopus 로고
    • The iron-regulated metastasis suppressor NDRG1 targets NEDD4L, PTEN, and SMAD4 and inhibits the PI3K and Ras signaling pathways
    • Kovacevic Z, Chikhani S, Lui GY, Sivagurunathan S, Richardson DR. The iron-regulated metastasis suppressor NDRG1 targets NEDD4L, PTEN, and SMAD4 and inhibits the PI3K and Ras signaling pathways. Antioxid Redox Signal. 2013; 18:874-887.
    • (2013) Antioxid Redox Signal. , vol.18 , pp. 874-887
    • Kovacevic, Z.1    Chikhani, S.2    Lui, G.Y.3    Sivagurunathan, S.4    Richardson, D.R.5
  • 188
    • 0035933726 scopus 로고    scopus 로고
    • Hypoxia induces the activation of the phosphatidylinositol 3-kinase/Akt cell survival pathway in PC12 cells: protective role in apoptosis
    • Alvarez-Tejado M, Naranjo-Suarez S, Jimenez C, Carrera AC, Landazuri MO, del Peso L. Hypoxia induces the activation of the phosphatidylinositol 3-kinase/Akt cell survival pathway in PC12 cells: protective role in apoptosis. J Biol Chem. 2001; 276:22368-22374.
    • (2001) J Biol Chem. , vol.276 , pp. 22368-22374
    • Alvarez-Tejado, M.1    Naranjo-Suarez, S.2    Jimenez, C.3    Carrera, A.C.4    Landazuri, M.O.5    del Peso, L.6
  • 189
    • 54249118338 scopus 로고    scopus 로고
    • Signaling and apoptosis differences between severe hypoxia and desferoxamine treatment of human epithelial cells
    • Box AH, Yuen C, Ponjevic D, Fick GH, Demetrick DJ. Signaling and apoptosis differences between severe hypoxia and desferoxamine treatment of human epithelial cells. Biochem Cell Biol. 2008; 86:425-436.
    • (2008) Biochem Cell Biol. , vol.86 , pp. 425-436
    • Box, A.H.1    Yuen, C.2    Ponjevic, D.3    Fick, G.H.4    Demetrick, D.J.5
  • 190
    • 40449120460 scopus 로고    scopus 로고
    • Iron depletion by deferoxamine up-regulates glucose uptake and insulin signaling in hepatoma cells and in rat liver
    • Dongiovanni P, Valenti L, Fracanzani AL, Gatti S, Cairo G, Fargion S. Iron depletion by deferoxamine up-regulates glucose uptake and insulin signaling in hepatoma cells and in rat liver. Am J Pathol. 2008; 172:738-747.
    • (2008) Am J Pathol. , vol.172 , pp. 738-747
    • Dongiovanni, P.1    Valenti, L.2    Fracanzani, A.L.3    Gatti, S.4    Cairo, G.5    Fargion, S.6
  • 191
    • 34247373460 scopus 로고    scopus 로고
    • Iron chelation regulates cyclin D1 expression via the proteasome: a link to iron deficiency-mediated growth suppression
    • Nurtjahja-Tjendraputra E, Fu D, Phang JM, Richardson DR. Iron chelation regulates cyclin D1 expression via the proteasome: a link to iron deficiency-mediated growth suppression. Blood. 2007; 109:4045-4054.
    • (2007) Blood. , vol.109 , pp. 4045-4054
    • Nurtjahja-Tjendraputra, E.1    Fu, D.2    Phang, J.M.3    Richardson, D.R.4
  • 192
    • 79958026380 scopus 로고    scopus 로고
    • The Ras-ERK and PI3K-mTOR pathways: cross-talk and compensation
    • Mendoza MC, Er EE, Blenis J. The Ras-ERK and PI3K-mTOR pathways: cross-talk and compensation. Trends Biochem Sci. 2011; 36:320-328.
    • (2011) Trends Biochem Sci. , vol.36 , pp. 320-328
    • Mendoza, M.C.1    Er, E.E.2    Blenis, J.3
  • 193
    • 84888132688 scopus 로고    scopus 로고
    • Signaling interplay between transforming growth factor-beta receptor and PI3K/AKT pathways in cancer
    • Zhang L, Zhou F, ten Dijke P. Signaling interplay between transforming growth factor-beta receptor and PI3K/AKT pathways in cancer. Trends Biochem Sci. 2013; 38:612-620.
    • (2013) Trends Biochem Sci. , vol.38 , pp. 612-620
    • Zhang, L.1    Zhou, F.2    ten Dijke, P.3
  • 194
    • 34248591612 scopus 로고    scopus 로고
    • Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer
    • Roberts PJ, Der CJ. Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene. 2007; 26:3291-3310.
    • (2007) Oncogene. , vol.26 , pp. 3291-3310
    • Roberts, P.J.1    Der, C.J.2
  • 195
    • 0037810249 scopus 로고    scopus 로고
    • Signal transduction mediated by the Ras/Raf/MEK/ERK pathway from cytokine receptors to transcription factors: potential targeting for therapeutic intervention
    • Chang F, Steelman LS, Lee JT, Shelton JG, Navolanic PM, Blalock WL, Franklin RA, McCubrey JA. Signal transduction mediated by the Ras/Raf/MEK/ERK pathway from cytokine receptors to transcription factors: potential targeting for therapeutic intervention. Leukemia. 2003; 17:1263-1293.
    • (2003) Leukemia. , vol.17 , pp. 1263-1293
    • Chang, F.1    Steelman, L.S.2    Lee, J.T.3    Shelton, J.G.4    Navolanic, P.M.5    Blalock, W.L.6    Franklin, R.A.7    McCubrey, J.A.8
  • 196
    • 2342591455 scopus 로고    scopus 로고
    • The discovery of receptor tyrosine kinases: targets for cancer therapy
    • Gschwind A, Fischer OM, Ullrich A. The discovery of receptor tyrosine kinases: targets for cancer therapy. Nat Rev Cancer. 2004; 4:361-370.
    • (2004) Nat Rev Cancer. , vol.4 , pp. 361-370
    • Gschwind, A.1    Fischer, O.M.2    Ullrich, A.3
  • 197
    • 25844457433 scopus 로고    scopus 로고
    • The epidermal growth factor receptor family
    • Bazley LA, Gullick WJ. The epidermal growth factor receptor family. Endocr Relat Cancer. 2005; 12:S17-S27.
    • (2005) Endocr Relat Cancer. , vol.12 , pp. S17-S27
    • Bazley, L.A.1    Gullick, W.J.2
  • 198
    • 0037145061 scopus 로고    scopus 로고
    • Ligand-induced, receptor-mediated dimerization and activation of EGF receptor
    • Schlessinger J. Ligand-induced, receptor-mediated dimerization and activation of EGF receptor. Cell. 2002; 110:669-672.
    • (2002) Cell. , vol.110 , pp. 669-672
    • Schlessinger, J.1
  • 200
    • 34547179997 scopus 로고    scopus 로고
    • MAP kinase pathways: the first twenty years
    • Avruch J. MAP kinase pathways: the first twenty years. Biochim Biophys Acta. 2007; 1773:1150-1160.
    • (2007) Biochim Biophys Acta. , vol.1773 , pp. 1150-1160
    • Avruch, J.1
  • 201
    • 0020315258 scopus 로고
    • Human genome contains four genes homologous to transforming genes of Harvey and Kirsten murine sarcoma viruses
    • Chang EH, Gonda MA, Ellis RW, Scolnick EM, Lowy DR. Human genome contains four genes homologous to transforming genes of Harvey and Kirsten murine sarcoma viruses. Proc Natl Acad Sci U S A. 1982; 79:4848-4852.
    • (1982) Proc Natl Acad Sci U S A. , vol.79 , pp. 4848-4852
    • Chang, E.H.1    Gonda, M.A.2    Ellis, R.W.3    Scolnick, E.M.4    Lowy, D.R.5
  • 202
    • 0037805547 scopus 로고    scopus 로고
    • RAS oncogenes: the first 30 years
    • Malumbres M, Barbacid M. RAS oncogenes: the first 30 years. Nat Rev Cancer. 2003; 3:459-465.
    • (2003) Nat Rev Cancer. , vol.3 , pp. 459-465
    • Malumbres, M.1    Barbacid, M.2
  • 203
    • 0030584088 scopus 로고    scopus 로고
    • Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-X(L)
    • Zha J, Harada H, Yang E, Jockel J, Korsmeyer SJ. Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-X(L). Cell. 1996; 87:619-628.
    • (1996) Cell. , vol.87 , pp. 619-628
    • Zha, J.1    Harada, H.2    Yang, E.3    Jockel, J.4    Korsmeyer, S.J.5
  • 204
    • 7444243152 scopus 로고    scopus 로고
    • Survival factor-induced extracellular signal-regulated kinase phosphorylates BIM, inhibiting its association with BAX and proapoptotic activity
    • Harada H, Quearry B, Ruiz-Vela A, Korsmeyer SJ. Survival factor-induced extracellular signal-regulated kinase phosphorylates BIM, inhibiting its association with BAX and proapoptotic activity. Proc Natl Acad Sci U S A. 2004; 101:15313-15317.
    • (2004) Proc Natl Acad Sci U S A. , vol.101 , pp. 15313-15317
    • Harada, H.1    Quearry, B.2    Ruiz-Vela, A.3    Korsmeyer, S.J.4
  • 205
    • 0038482050 scopus 로고    scopus 로고
    • Activation of the ERK1/2 signaling pathway promotes phosphorylation and proteasome-dependent degradation of the BH3-only protein, Bim
    • Ley R, Balmanno K, Hadfield K, Weston C, Cook SJ. Activation of the ERK1/2 signaling pathway promotes phosphorylation and proteasome-dependent degradation of the BH3-only protein, Bim. J Biol Chem. 2003; 278:18811-18816.
    • (2003) J Biol Chem. , vol.278 , pp. 18811-18816
    • Ley, R.1    Balmanno, K.2    Hadfield, K.3    Weston, C.4    Cook, S.J.5
  • 208
    • 0037264633 scopus 로고    scopus 로고
    • Targeting RAS signalling pathways in cancer therapy
    • Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003; 3:11-22.
    • (2003) Nat Rev Cancer. , vol.3 , pp. 11-22
    • Downward, J.1
  • 209
    • 34248583886 scopus 로고    scopus 로고
    • MAP kinase signalling pathways in cancer
    • Dhillon AS, Hagan S, Rath O, Kolch W. MAP kinase signalling pathways in cancer. Oncogene. 2007; 26:3279-3290.
    • (2007) Oncogene. , vol.26 , pp. 3279-3290
    • Dhillon, A.S.1    Hagan, S.2    Rath, O.3    Kolch, W.4
  • 210
    • 34347395733 scopus 로고    scopus 로고
    • Trastuzumab-mechanism of action and use in clinical practice
    • Hudis CA. Trastuzumab-mechanism of action and use in clinical practice. N Engl J Med. 2007; 357:39-51.
    • (2007) N Engl J Med. , vol.357 , pp. 39-51
    • Hudis, C.A.1
  • 211
    • 84888196929 scopus 로고    scopus 로고
    • Pharmacokinetics, clinical indications, and resistance mechanisms in molecular targeted therapies in cancer
    • Izar B, Rotow J, Gainor J, Clark J, Chabner B. Pharmacokinetics, clinical indications, and resistance mechanisms in molecular targeted therapies in cancer. Pharmacol Rev. 2013; 65:1351-1395.
    • (2013) Pharmacol Rev. , vol.65 , pp. 1351-1395
    • Izar, B.1    Rotow, J.2    Gainor, J.3    Clark, J.4    Chabner, B.5
  • 214
    • 0035282334 scopus 로고    scopus 로고
    • Mammalian MAP kinase signalling cascades
    • Chang L, Karin M. Mammalian MAP kinase signalling cascades. Nature. 2001; 410:37-40.
    • (2001) Nature. , vol.410 , pp. 37-40
    • Chang, L.1    Karin, M.2
  • 215
    • 84890447356 scopus 로고    scopus 로고
    • Role of p38 MAP kinase signal transduction in solid tumors
    • Koul HK, Pal M, Koul S. Role of p38 MAP kinase signal transduction in solid tumors. Genes Cancer. 2013; 4:342-359.
    • (2013) Genes Cancer. , vol.4 , pp. 342-359
    • Koul, H.K.1    Pal, M.2    Koul, S.3
  • 216
    • 4444240191 scopus 로고    scopus 로고
    • The functional interactions between the p53 and MAPK signaling pathways
    • Wu GS. The functional interactions between the p53 and MAPK signaling pathways. Cancer Biol Ther. 2004; 3:156-161.
    • (2004) Cancer Biol Ther. , vol.3 , pp. 156-161
    • Wu, G.S.1
  • 217
    • 67749117934 scopus 로고    scopus 로고
    • Signal integration by JNK and p38 MAPK pathways in cancer development
    • Wagner EF, Nebreda AR. Signal integration by JNK and p38 MAPK pathways in cancer development. Nat Rev Cancer. 2009; 9:537-549.
    • (2009) Nat Rev Cancer. , vol.9 , pp. 537-549
    • Wagner, E.F.1    Nebreda, A.R.2
  • 219
    • 33646814635 scopus 로고    scopus 로고
    • Targeted deletion of MKK4 in cancer cells: a detrimental phenotype manifests as decreased experimental metastasis and suggests a counterweight to the evolution of tumor-suppressor loss
    • Cunningham SC, Gallmeier E, Hucl T, Dezentje DA, Calhoun ES, Falco G, Abdelmohsen K, Gorospe M, Kern SE. Targeted deletion of MKK4 in cancer cells: a detrimental phenotype manifests as decreased experimental metastasis and suggests a counterweight to the evolution of tumor-suppressor loss. Cancer Res. 2006; 66:5560-5564.
    • (2006) Cancer Res. , vol.66 , pp. 5560-5564
    • Cunningham, S.C.1    Gallmeier, E.2    Hucl, T.3    Dezentje, D.A.4    Calhoun, E.S.5    Falco, G.6    Abdelmohsen, K.7    Gorospe, M.8    Kern, S.E.9
  • 220
    • 40849145457 scopus 로고    scopus 로고
    • Desferrioxamine (DFX) induces apoptosis through the p38-caspase8-Bid-Bax pathway in PHA-stimulated human lymphocytes
    • Kim BM, Chung HW. Desferrioxamine (DFX) induces apoptosis through the p38-caspase8-Bid-Bax pathway in PHA-stimulated human lymphocytes. Toxicol Appl Pharmacol. 2008; 228:24-31.
    • (2008) Toxicol Appl Pharmacol. , vol.228 , pp. 24-31
    • Kim, B.M.1    Chung, H.W.2
  • 221
    • 79955462543 scopus 로고    scopus 로고
    • Cellular iron depletion stimulates the JNK and p38 MAPK signaling transduction pathways, dissociation of ASK1-thioredoxin, and activation of ASK1
    • Yu Y, Richardson DR. Cellular iron depletion stimulates the JNK and p38 MAPK signaling transduction pathways, dissociation of ASK1-thioredoxin, and activation of ASK1. J Biol Chem. 2011; 286:15413-15427.
    • (2011) J Biol Chem. , vol.286 , pp. 15413-15427
    • Yu, Y.1    Richardson, D.R.2
  • 223
    • 33747359592 scopus 로고    scopus 로고
    • p38 and ERK MAP kinase mediates iron chelator-induced apoptosis and -suppressed differentiation of immortalized and malignant human oral keratinocytes
    • Lee SK, Jang HJ, Lee HJ, Lee J, Jeon BH, Jun CD, Lee SK, Kim EC. p38 and ERK MAP kinase mediates iron chelator-induced apoptosis and -suppressed differentiation of immortalized and malignant human oral keratinocytes. Life Sci. 2006; 79:1419-1427.
    • (2006) Life Sci. , vol.79 , pp. 1419-1427
    • Lee, S.K.1    Jang, H.J.2    Lee, H.J.3    Lee, J.4    Jeon, B.H.5    Jun, C.D.6    Lee, S.K.7    Kim, E.C.8
  • 225
    • 0037200121 scopus 로고    scopus 로고
    • Raf-independent deregulation of p38 and JNK mitogen-activated protein kinases are critical for Ras transformation
    • Pruitt K, Pruitt WM, Bilter GK, Westwick JK, Der CJ. Raf-independent deregulation of p38 and JNK mitogen-activated protein kinases are critical for Ras transformation. J Biol Chem. 2002; 277:31808-31817.
    • (2002) J Biol Chem. , vol.277 , pp. 31808-31817
    • Pruitt, K.1    Pruitt, W.M.2    Bilter, G.K.3    Westwick, J.K.4    Der, C.J.5
  • 226
    • 35048875855 scopus 로고    scopus 로고
    • TGF-beta signaling: A tale of two responses
    • Rahimi RA, Leof EB. TGF-beta signaling: A tale of two responses. J Cell Biochem. 2007; 102:593-608.
    • (2007) J Cell Biochem. , vol.102 , pp. 593-608
    • Rahimi, R.A.1    Leof, E.B.2
  • 227
    • 0030613249 scopus 로고    scopus 로고
    • T beta RI phosphorylation of Smad2 on Ser(465) and Ser(467) is required for Smad2-Smad4 complex formation and signaling
    • Abdollah S, MaciasSilva M, Tsukazaki T, Hayashi H, Attisano L, Wrana JL. T beta RI phosphorylation of Smad2 on Ser(465) and Ser(467) is required for Smad2-Smad4 complex formation and signaling. J Biol Chem. 1997; 272:27678-27685.
    • (1997) J Biol Chem. , vol.272 , pp. 27678-27685
    • Abdollah, S.1    MaciasSilva, M.2    Tsukazaki, T.3    Hayashi, H.4    Attisano, L.5    Wrana, J.L.6
  • 228
    • 0030613262 scopus 로고    scopus 로고
    • Phosphorylation of Ser(465) and Ser(467) in the C terminus of Smad2 mediates interaction with Smad4 and is required for transforming growth factor-beta signaling
    • Souchelnytskyi S, Tamaki K, Engstrom U, Wernstedt C, tenDijke P, Heldin CH. Phosphorylation of Ser(465) and Ser(467) in the C terminus of Smad2 mediates interaction with Smad4 and is required for transforming growth factor-beta signaling. J Biol Chem. 1997; 272:28107-28115.
    • (1997) J Biol Chem. , vol.272 , pp. 28107-28115
    • Souchelnytskyi, S.1    Tamaki, K.2    Engstrom, U.3    Wernstedt, C.4    tenDijke, P.5    Heldin, C.H.6
  • 230
    • 0034644472 scopus 로고    scopus 로고
    • TGF beta signaling in growth control, cancer, and heritable disorders
    • Massague J, Blain SW, Lo RS. TGF beta signaling in growth control, cancer, and heritable disorders. Cell. 2000; 103:295-309.
    • (2000) Cell. , vol.103 , pp. 295-309
    • Massague, J.1    Blain, S.W.2    Lo, R.S.3
  • 231
    • 0033559189 scopus 로고    scopus 로고
    • Restoration of transforming growth factor beta signaling pathway in human prostate cancer cells suppresses tumorigenicity via induction of caspase-1-mediated apoptosis
    • Guo YP, Kyprianou N. Restoration of transforming growth factor beta signaling pathway in human prostate cancer cells suppresses tumorigenicity via induction of caspase-1-mediated apoptosis. Cancer Res. 1999; 59:1366-1371.
    • (1999) Cancer Res. , vol.59 , pp. 1366-1371
    • Guo, Y.P.1    Kyprianou, N.2
  • 232
    • 0026667739 scopus 로고
    • Interaction of epidermal growth factor and transforming growth factor-beta in human prostatic epithelial cells in culture
    • Sutkowski DM, Fong CJ, Sensibar JA, Rademaker AW, Sherwood ER, Kozlowski JM, Chung L. Interaction of epidermal growth factor and transforming growth factor-beta in human prostatic epithelial cells in culture. Prostate. 1992; 21:133-143.
    • (1992) Prostate. , vol.21 , pp. 133-143
    • Sutkowski, D.M.1    Fong, C.J.2    Sensibar, J.A.3    Rademaker, A.W.4    Sherwood, E.R.5    Kozlowski, J.M.6    Chung, L.7
  • 233
    • 0026342978 scopus 로고
    • Response of prostate cancer cells to peptide growth factors - transforming growth factor-beta
    • Wilding G. Response of prostate cancer cells to peptide growth factors - transforming growth factor-beta. Cancer Surv. 1991; 11:147-163.
    • (1991) Cancer Surv. , vol.11 , pp. 147-163
    • Wilding, G.1
  • 234
    • 0033106484 scopus 로고    scopus 로고
    • A mechanism of repression of TGF beta/Smad signaling by oncogenic Ras
    • Kretzschmar M, Doody J, Timokhina I, Massague J. A mechanism of repression of TGF beta/Smad signaling by oncogenic Ras. Genes Dev. 1999; 13:804-816.
    • (1999) Genes Dev. , vol.13 , pp. 804-816
    • Kretzschmar, M.1    Doody, J.2    Timokhina, I.3    Massague, J.4
  • 235
    • 0028953810 scopus 로고
    • Transforming growth-factor-beta and cell-cycle regulation
    • Alexandrow MG, Moses HL. Transforming growth-factor-beta and cell-cycle regulation. Cancer Res. 1995; 55:1452-1457.
    • (1995) Cancer Res. , vol.55 , pp. 1452-1457
    • Alexandrow, M.G.1    Moses, H.L.2
  • 236
    • 84886496878 scopus 로고    scopus 로고
    • Role of STAT3 in cancer metastasis and translational advances
    • Kamran MZ, Patil P, Gude RP. Role of STAT3 in cancer metastasis and translational advances. Biomed Res Int. 2013; 2013:421821.
    • (2013) Biomed Res Int. , vol.2013 , pp. 421821
    • Kamran, M.Z.1    Patil, P.2    Gude, R.P.3
  • 237
    • 0028845212 scopus 로고
    • Cytokine receptor signalling
    • Ihle JN. Cytokine receptor signalling. Nature. 1995; 377:591-594.
    • (1995) Nature. , vol.377 , pp. 591-594
    • Ihle, J.N.1
  • 238
    • 0028349735 scopus 로고
    • Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6
    • Zhong Z, Wen Z, Darnell JE Jr,. Stat3: a STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6. Science. 1994; 264:95-98.
    • (1994) Science. , vol.264 , pp. 95-98
    • Zhong, Z.1    Wen, Z.2    Darnell, J.E.3
  • 239
    • 0029069540 scopus 로고
    • Enhanced DNA-binding activity of a Stat3-related protein in cells transformed by the Src oncoprotein
    • Yu CL, Meyer DJ, Campbell GS, Larner AC, Carter-Su C, Schwartz J, Jove R. Enhanced DNA-binding activity of a Stat3-related protein in cells transformed by the Src oncoprotein. Science. 1995; 269:81-83.
    • (1995) Science. , vol.269 , pp. 81-83
    • Yu, C.L.1    Meyer, D.J.2    Campbell, G.S.3    Larner, A.C.4    Carter-Su, C.5    Schwartz, J.6    Jove, R.7
  • 240
    • 0034722896 scopus 로고    scopus 로고
    • STAT proteins: novel molecular targets for cancer drug discovery
    • Turkson J, Jove R. STAT proteins: novel molecular targets for cancer drug discovery. Oncogene. 2000; 19:6613-6626.
    • (2000) Oncogene. , vol.19 , pp. 6613-6626
    • Turkson, J.1    Jove, R.2
  • 241
    • 0033451992 scopus 로고    scopus 로고
    • Synergistic roles for Pim-1 and c-Myc in STAT3-mediated cell cycle progression and antiapoptosis
    • Shirogane T, Fukada T, Muller JM, Shima DT, Hibi M, Hirano T. Synergistic roles for Pim-1 and c-Myc in STAT3-mediated cell cycle progression and antiapoptosis. Immunity. 1999; 11:709-719.
    • (1999) Immunity. , vol.11 , pp. 709-719
    • Shirogane, T.1    Fukada, T.2    Muller, J.M.3    Shima, D.T.4    Hibi, M.5    Hirano, T.6
  • 243
    • 2442685645 scopus 로고    scopus 로고
    • Stat3 activation regulates the expression of matrix metalloproteinase-2 and tumor invasion and metastasis
    • Xie TX, Wei D, Liu M, Gao AC, Ali-Osman F, Sawaya R, Huang S. Stat3 activation regulates the expression of matrix metalloproteinase-2 and tumor invasion and metastasis. Oncogene. 2004; 23:3550-3560.
    • (2004) Oncogene. , vol.23 , pp. 3550-3560
    • Xie, T.X.1    Wei, D.2    Liu, M.3    Gao, A.C.4    Ali-Osman, F.5    Sawaya, R.6    Huang, S.7
  • 244
    • 20444453281 scopus 로고    scopus 로고
    • A role of STAT3 in Rho GTPase-regulated cell migration and proliferation
    • Debidda M, Wang L, Zang H, Poli V, Zheng Y. A role of STAT3 in Rho GTPase-regulated cell migration and proliferation. J Biol Chem. 2005; 280:17275-17285.
    • (2005) J Biol Chem. , vol.280 , pp. 17275-17285
    • Debidda, M.1    Wang, L.2    Zang, H.3    Poli, V.4    Zheng, Y.5
  • 248
    • 84860630975 scopus 로고    scopus 로고
    • Targeting the interleukin-6/Jak/Stat pathway in human malignancies
    • Sansone P, Bromberg J. Targeting the interleukin-6/Jak/Stat pathway in human malignancies. J Clin Oncol. 2012; 30:1005-1014.
    • (2012) J Clin Oncol. , vol.30 , pp. 1005-1014
    • Sansone, P.1    Bromberg, J.2
  • 250
    • 0030772458 scopus 로고    scopus 로고
    • STAT3 serine phosphorylation by ERK-dependent and -independent pathways negatively modulates its tyrosine phosphorylation
    • Chung J, Uchida E, Grammer TC, Blenis J. STAT3 serine phosphorylation by ERK-dependent and -independent pathways negatively modulates its tyrosine phosphorylation. Mol Cell Biol. 1997; 17:6508-6516.
    • (1997) Mol Cell Biol. , vol.17 , pp. 6508-6516
    • Chung, J.1    Uchida, E.2    Grammer, T.C.3    Blenis, J.4
  • 251
    • 0032542211 scopus 로고    scopus 로고
    • Repression of Stat3 activity by activation of mitogen-activated protein kinase (MAPK)
    • Jain N, Zhang T, Fong SL, Lim CP, Cao X. Repression of Stat3 activity by activation of mitogen-activated protein kinase (MAPK). Oncogene. 1998; 17:3157-3167.
    • (1998) Oncogene. , vol.17 , pp. 3157-3167
    • Jain, N.1    Zhang, T.2    Fong, S.L.3    Lim, C.P.4    Cao, X.5
  • 253
    • 0035877595 scopus 로고    scopus 로고
    • Regulation of Stat3 activation by MEK kinase 1
    • Lim CP, Cao X. Regulation of Stat3 activation by MEK kinase 1. J Biol Chem. 2001; 276:21004-21011.
    • (2001) J Biol Chem. , vol.276 , pp. 21004-21011
    • Lim, C.P.1    Cao, X.2
  • 254
    • 1642299884 scopus 로고    scopus 로고
    • Cytoplasmic c-Fos induced by the YXXQ-derived STAT3 signal requires the co-operative MEK/ERK signal for its nuclear translocation
    • Higashi N, Kunimoto H, Kaneko S, Sasaki T, Ishii M, Kojima H, Nakajima K. Cytoplasmic c-Fos induced by the YXXQ-derived STAT3 signal requires the co-operative MEK/ERK signal for its nuclear translocation. Genes Cells. 2004; 9:233-242.
    • (2004) Genes Cells. , vol.9 , pp. 233-242
    • Higashi, N.1    Kunimoto, H.2    Kaneko, S.3    Sasaki, T.4    Ishii, M.5    Kojima, H.6    Nakajima, K.7
  • 255
    • 0036298183 scopus 로고    scopus 로고
    • PTEN is a negative regulator of STAT3 activation in human papillomavirus-infected cells
    • Sun S, Steinberg BM. PTEN is a negative regulator of STAT3 activation in human papillomavirus-infected cells. J Gen Virol. 2002; 83:1651-1658.
    • (2002) J Gen Virol. , vol.83 , pp. 1651-1658
    • Sun, S.1    Steinberg, B.M.2
  • 257
    • 84919329880 scopus 로고    scopus 로고
    • Integrated analysis reveals that STAT3 is central to the crosstalk between HER/ErbB receptor signaling pathways in human mammary epithelial cells
    • Gong C, Zhang Y, Shankaran H, Resat H. Integrated analysis reveals that STAT3 is central to the crosstalk between HER/ErbB receptor signaling pathways in human mammary epithelial cells. Mol Biosyst. 2015; 11:146-158.
    • (2015) Mol Biosyst. , vol.11 , pp. 146-158
    • Gong, C.1    Zhang, Y.2    Shankaran, H.3    Resat, H.4
  • 258
    • 84894593599 scopus 로고    scopus 로고
    • Molecular mechanisms of epithelial-mesenchymal transition
    • Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 2014; 15:178-196.
    • (2014) Nat Rev Mol Cell Biol. , vol.15 , pp. 178-196
    • Lamouille, S.1    Xu, J.2    Derynck, R.3
  • 261
    • 58149213801 scopus 로고    scopus 로고
    • Non-Smad pathways in TGF-beta signaling
    • Zhang YE. Non-Smad pathways in TGF-beta signaling. Cell Res. 2009; 19:128-139.
    • (2009) Cell Res. , vol.19 , pp. 128-139
    • Zhang, Y.E.1
  • 262
    • 0034214211 scopus 로고    scopus 로고
    • Mitogenic conversion of transforming growth factor-beta 1 effect by oncogenic Ha-Ras-induced activation of the mitogen-activated protein kinase signaling pathway in human prostate cancer
    • Park BJ, Park JI, Byun DS, Park JH, Chi SG. Mitogenic conversion of transforming growth factor-beta 1 effect by oncogenic Ha-Ras-induced activation of the mitogen-activated protein kinase signaling pathway in human prostate cancer. Cancer Res. 2000; 60:3031-3038.
    • (2000) Cancer Res. , vol.60 , pp. 3031-3038
    • Park, B.J.1    Park, J.I.2    Byun, D.S.3    Park, J.H.4    Chi, S.G.5
  • 264
    • 34548496683 scopus 로고    scopus 로고
    • Differential regulation of epithelial and mesenchymal markers by deltaEF1 proteins in epithelial mesenchymal transition induced by TGF-beta
    • Shirakihara T, Saitoh M, Miyazono K. Differential regulation of epithelial and mesenchymal markers by deltaEF1 proteins in epithelial mesenchymal transition induced by TGF-beta. Mol Biol Cell. 2007; 18:3533-3544.
    • (2007) Mol Biol Cell. , vol.18 , pp. 3533-3544
    • Shirakihara, T.1    Saitoh, M.2    Miyazono, K.3
  • 265
    • 0038369998 scopus 로고    scopus 로고
    • A self-enabling TGFbeta response coupled to stress signaling: Smad engages stress response factor ATF3 for Id1 repression in epithelial cells
    • Kang Y, Chen CR, Massague J. A self-enabling TGFbeta response coupled to stress signaling: Smad engages stress response factor ATF3 for Id1 repression in epithelial cells. Mol Cell. 2003; 11:915-926.
    • (2003) Mol Cell. , vol.11 , pp. 915-926
    • Kang, Y.1    Chen, C.R.2    Massague, J.3
  • 266
    • 0028603343 scopus 로고
    • TGF-beta induced transdifferentiation of mammary epithelial cells to mesenchymal cells: involvement of type I receptors
    • Miettinen PJ, Ebner R, Lopez AR, Derynck R. TGF-beta induced transdifferentiation of mammary epithelial cells to mesenchymal cells: involvement of type I receptors. J Cell Biol. 1994; 127:2021-2036.
    • (1994) J Cell Biol. , vol.127 , pp. 2021-2036
    • Miettinen, P.J.1    Ebner, R.2    Lopez, A.R.3    Derynck, R.4
  • 267
    • 78650510746 scopus 로고    scopus 로고
    • Emergence of the phosphoinositide 3-kinase-Akt-mammalian target of rapamycin axis in transforming growth factor-beta-induced epithelial-mesenchymal transition
    • Lamouille S, Derynck R. Emergence of the phosphoinositide 3-kinase-Akt-mammalian target of rapamycin axis in transforming growth factor-beta-induced epithelial-mesenchymal transition. Cells Tissues Organs. 2011; 193:8-22.
    • (2011) Cells Tissues Organs. , vol.193 , pp. 8-22
    • Lamouille, S.1    Derynck, R.2
  • 268
    • 84871234148 scopus 로고    scopus 로고
    • TWIST1 is an ERK1/2 effector that promotes invasion and regulates MMP-1 expression in human melanoma cells
    • Weiss MB, Abel EV, Mayberry MM, Basile KJ, Berger AC, Aplin AE. TWIST1 is an ERK1/2 effector that promotes invasion and regulates MMP-1 expression in human melanoma cells. Cancer Res. 2012; 72:6382-6392.
    • (2012) Cancer Res. , vol.72 , pp. 6382-6392
    • Weiss, M.B.1    Abel, E.V.2    Mayberry, M.M.3    Basile, K.J.4    Berger, A.C.5    Aplin, A.E.6
  • 269
    • 42249109021 scopus 로고    scopus 로고
    • Synergistic effect between EGF and TGF-beta1 in inducing oncogenic properties of intestinal epithelial cells
    • Uttamsingh S, Bao X, Nguyen KT, Bhanot M, Gong J, Chan JL, Liu F, Chu TT, Wang LH. Synergistic effect between EGF and TGF-beta1 in inducing oncogenic properties of intestinal epithelial cells. Oncogene. 2008; 27:2626-2634.
    • (2008) Oncogene. , vol.27 , pp. 2626-2634
    • Uttamsingh, S.1    Bao, X.2    Nguyen, K.T.3    Bhanot, M.4    Gong, J.5    Chan, J.L.6    Liu, F.7    Chu, T.T.8    Wang, L.H.9
  • 270
    • 34547587877 scopus 로고    scopus 로고
    • Cell size and invasion in TGF-beta-induced epithelial to mesenchymal transition is regulated by activation of the mTOR pathway
    • Lamouille S, Derynck R. Cell size and invasion in TGF-beta-induced epithelial to mesenchymal transition is regulated by activation of the mTOR pathway. J Cell Biol. 2007; 178:437-451.
    • (2007) J Cell Biol. , vol.178 , pp. 437-451
    • Lamouille, S.1    Derynck, R.2
  • 271
    • 84861448736 scopus 로고    scopus 로고
    • TGF-beta-induced activation of mTOR complex 2 drives epithelial-mesenchymal transition and cell invasion
    • Lamouille S, Connolly E, Smyth JW, Akhurst RJ, Derynck R. TGF-beta-induced activation of mTOR complex 2 drives epithelial-mesenchymal transition and cell invasion. J Cell Sci. 2012; 125:1259-1273.
    • (2012) J Cell Sci. , vol.125 , pp. 1259-1273
    • Lamouille, S.1    Connolly, E.2    Smyth, J.W.3    Akhurst, R.J.4    Derynck, R.5
  • 272
    • 12144266241 scopus 로고    scopus 로고
    • Glycogen synthase kinase-3 is an endogenous inhibitor of Snail transcription: implications for the epithelial-mesenchymal transition
    • Bachelder RE, Yoon SO, Franci C, de Herreros AG, Mercurio AM. Glycogen synthase kinase-3 is an endogenous inhibitor of Snail transcription: implications for the epithelial-mesenchymal transition. J Cell Biol. 2005; 168:29-33.
    • (2005) J Cell Biol. , vol.168 , pp. 29-33
    • Bachelder, R.E.1    Yoon, S.O.2    Franci, C.3    de Herreros, A.G.4    Mercurio, A.M.5
  • 275
    • 15744394838 scopus 로고    scopus 로고
    • LIM-kinase 2 and cofilin phosphorylation mediate actin cytoskeleton reorganization induced by transforming growth factor-beta
    • Vardouli L, Moustakas A, Stournaras C. LIM-kinase 2 and cofilin phosphorylation mediate actin cytoskeleton reorganization induced by transforming growth factor-beta. J Biol Chem. 2005; 280:11448-11457.
    • (2005) J Biol Chem. , vol.280 , pp. 11448-11457
    • Vardouli, L.1    Moustakas, A.2    Stournaras, C.3
  • 276
    • 8444251784 scopus 로고    scopus 로고
    • The Wnt signaling pathway in development and disease
    • Logan CY, Nusse R. The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol. 2004; 20:781-810.
    • (2004) Annu Rev Cell Dev Biol. , vol.20 , pp. 781-810
    • Logan, C.Y.1    Nusse, R.2
  • 277
    • 77954210220 scopus 로고    scopus 로고
    • An updated overview on Wnt signaling pathways: a prelude for more
    • Rao TP, Kuhl M. An updated overview on Wnt signaling pathways: a prelude for more. Circ Res. 2010; 106:1798-1806.
    • (2010) Circ Res. , vol.106 , pp. 1798-1806
    • Rao, T.P.1    Kuhl, M.2
  • 279
    • 84867371728 scopus 로고    scopus 로고
    • Canonical Wnt signaling regulates Slug activity and links epithelial-mesenchymal transition with epigenetic Breast Cancer 1, Early Onset (BRCA1) repression
    • Wu ZQ, Li XY, Hu CY, Ford M, Kleer CG, Weiss SJ. Canonical Wnt signaling regulates Slug activity and links epithelial-mesenchymal transition with epigenetic Breast Cancer 1, Early Onset (BRCA1) repression. Proc Natl Acad Sci U S A. 2012; 109:16654-16659.
    • (2012) Proc Natl Acad Sci U S A. , vol.109 , pp. 16654-16659
    • Wu, Z.Q.1    Li, X.Y.2    Hu, C.Y.3    Ford, M.4    Kleer, C.G.5    Weiss, S.J.6
  • 280
    • 84908207903 scopus 로고    scopus 로고
    • N-myc downstream-regulated gene 1 downregulates cell proliferation, invasiveness, and tumorigenesis in human oral squamous cell carcinoma
    • Lee JC, Chung LC, Chen YJ, Feng TH, Juang HH. N-myc downstream-regulated gene 1 downregulates cell proliferation, invasiveness, and tumorigenesis in human oral squamous cell carcinoma. Cancer Lett. 2014; 355:242-252.
    • (2014) Cancer Lett. , vol.355 , pp. 242-252
    • Lee, J.C.1    Chung, L.C.2    Chen, Y.J.3    Feng, T.H.4    Juang, H.H.5
  • 281
    • 84904350451 scopus 로고    scopus 로고
    • The metastasis suppressor NDRG1 modulates the phosphorylation and nuclear translocation of beta-catenin through mechanisms involving FRAT1 and PAK4
    • Jin R, Liu W, Menezes S, Yue F, Zheng M, Kovacevic Z, Richardson DR. The metastasis suppressor NDRG1 modulates the phosphorylation and nuclear translocation of beta-catenin through mechanisms involving FRAT1 and PAK4. J Cell Sci. 2014; 127:3116-3130.
    • (2014) J Cell Sci. , vol.127 , pp. 3116-3130
    • Jin, R.1    Liu, W.2    Menezes, S.3    Yue, F.4    Zheng, M.5    Kovacevic, Z.6    Richardson, D.R.7
  • 284
    • 84891948851 scopus 로고    scopus 로고
    • Deferoxamine enhances cell migration and invasion through promotion of HIF-1alpha expression and epithelial-mesenchymal transition in colorectal cancer
    • Zhang W, Wu Y, Yan Q, Ma F, Shi X, Zhao Y, Peng Y, Wang J, Jiang B. Deferoxamine enhances cell migration and invasion through promotion of HIF-1alpha expression and epithelial-mesenchymal transition in colorectal cancer. Oncol Rep. 2014; 31:111-116.
    • (2014) Oncol Rep. , vol.31 , pp. 111-116
    • Zhang, W.1    Wu, Y.2    Yan, Q.3    Ma, F.4    Shi, X.5    Zhao, Y.6    Peng, Y.7    Wang, J.8    Jiang, B.9
  • 286
    • 84863113109 scopus 로고    scopus 로고
    • Actin stress fibers-assembly, dynamics and biological roles
    • Tojkander S, Gateva G, Lappalainen P. Actin stress fibers-assembly, dynamics and biological roles. J Cell Sci. 2012; 125:1855-1864.
    • (2012) J Cell Sci. , vol.125 , pp. 1855-1864
    • Tojkander, S.1    Gateva, G.2    Lappalainen, P.3
  • 287
    • 84863767949 scopus 로고    scopus 로고
    • Functions of nonmuscle myosin II in assembly of the cellular contractile system
    • Shutova M, Yang C, Vasiliev JM, Svitkina T. Functions of nonmuscle myosin II in assembly of the cellular contractile system. PLoS One. 2012; 7:e40814.
    • (2012) PLoS One. , vol.7 , pp. e40814
    • Shutova, M.1    Yang, C.2    Vasiliev, J.M.3    Svitkina, T.4
  • 288
    • 10944244704 scopus 로고    scopus 로고
    • Rho signalling at a glance
    • Schwartz M. Rho signalling at a glance. J Cell Sci. 2004; 117:5457-5458.
    • (2004) J Cell Sci. , vol.117 , pp. 5457-5458
    • Schwartz, M.1
  • 289
    • 0842281652 scopus 로고    scopus 로고
    • Rho and Rac take center stage
    • Burridge K, Wennerberg K. Rho and Rac take center stage. Cell. 2004; 116:167-179.
    • (2004) Cell. , vol.116 , pp. 167-179
    • Burridge, K.1    Wennerberg, K.2
  • 290
    • 79251516379 scopus 로고    scopus 로고
    • Rho-associated kinase-dependent contraction of stress fibres and the organization of focal adhesions
    • Katoh K, Kano Y, Noda Y. Rho-associated kinase-dependent contraction of stress fibres and the organization of focal adhesions. J R Soc Interface. 2011; 8:305-311.
    • (2011) J R Soc Interface. , vol.8 , pp. 305-311
    • Katoh, K.1    Kano, Y.2    Noda, Y.3
  • 291
    • 0038386613 scopus 로고    scopus 로고
    • Src family kinases in tumor progression and metastasis
    • Summy JM, Gallick GE. Src family kinases in tumor progression and metastasis. Cancer Metastasis Rev. 2003; 22:337-358.
    • (2003) Cancer Metastasis Rev. , vol.22 , pp. 337-358
    • Summy, J.M.1    Gallick, G.E.2
  • 292
    • 1842584776 scopus 로고    scopus 로고
    • Newest findings on the oldest oncogene; how activated src does it
    • Frame MC. Newest findings on the oldest oncogene; how activated src does it. J Cell Sci. 2004; 117:989-998.
    • (2004) J Cell Sci. , vol.117 , pp. 989-998
    • Frame, M.C.1
  • 295
    • 12944308330 scopus 로고    scopus 로고
    • Eating oneself and uninvited guests: autophagy-related pathways in cellular defense
    • Levine B. Eating oneself and uninvited guests: autophagy-related pathways in cellular defense. Cell. 2005; 120:159-162.
    • (2005) Cell. , vol.120 , pp. 159-162
    • Levine, B.1
  • 296
    • 84863428905 scopus 로고    scopus 로고
    • Autophagy regulation and integration with cell signaling
    • Hamacher-Brady A. Autophagy regulation and integration with cell signaling. Antioxid Redox Signal. 2012; 17:756-765.
    • (2012) Antioxid Redox Signal. , vol.17 , pp. 756-765
    • Hamacher-Brady, A.1
  • 297
    • 77951214016 scopus 로고    scopus 로고
    • Mammalian autophagy: core molecular machinery and signaling regulation
    • Yang Z, Klionsky DJ. Mammalian autophagy: core molecular machinery and signaling regulation. Curr Opin Cell Biol. 2010; 22:124-131.
    • (2010) Curr Opin Cell Biol. , vol.22 , pp. 124-131
    • Yang, Z.1    Klionsky, D.J.2
  • 299
    • 80052697287 scopus 로고    scopus 로고
    • The role of autophagy in cancer: therapeutic implications
    • Yang ZJ, Chee CE, Huang S, Sinicrope FA. The role of autophagy in cancer: therapeutic implications. Mol Cancer Ther. 2011; 10:1533-1541.
    • (2011) Mol Cancer Ther. , vol.10 , pp. 1533-1541
    • Yang, Z.J.1    Chee, C.E.2    Huang, S.3    Sinicrope, F.A.4
  • 300
    • 84924762625 scopus 로고    scopus 로고
    • Iron metabolism and autophagy: a poorly explored relationship that has important consequences for health and disease
    • Krishan S, Jansson PJ, Gutierrez E, Lane DJ, Richardson DES, Sahni S. Iron metabolism and autophagy: a poorly explored relationship that has important consequences for health and disease. Nagoya J Med Sci. 2015; 77:1-6.
    • (2015) Nagoya J Med Sci. , vol.77 , pp. 1-6
    • Krishan, S.1    Jansson, P.J.2    Gutierrez, E.3    Lane, D.J.4    Richardson, D.E.S.5    Sahni, S.6
  • 302
    • 36248949141 scopus 로고    scopus 로고
    • The endoplasmic reticulum and the unfolded protein response
    • Malhotra JD, Kaufman RJ. The endoplasmic reticulum and the unfolded protein response. Semin Cell Dev Biol. 2007; 18:716-731.
    • (2007) Semin Cell Dev Biol. , vol.18 , pp. 716-731
    • Malhotra, J.D.1    Kaufman, R.J.2
  • 307
    • 20344387475 scopus 로고    scopus 로고
    • Autophagy: dual roles in life and death?
    • Baehrecke EH. Autophagy: dual roles in life and death? Nat Rev Mol Cell Biol. 2005; 6:505-510.
    • (2005) Nat Rev Mol Cell Biol , vol.6 , pp. 505-510
    • Baehrecke, E.H.1
  • 308
    • 84912098442 scopus 로고    scopus 로고
    • The anti-cancer agent, Dp44mT, overcomes pro-survival autophagy by two mechanisms: persistent induction of autophagosome synthesis and impairment of lysosomal integrity
    • Gutierrez E, Richardson DR, Jansson PJ. The anti-cancer agent, Dp44mT, overcomes pro-survival autophagy by two mechanisms: persistent induction of autophagosome synthesis and impairment of lysosomal integrity. J Biol Chem. 2014; 289:33568-33589.
    • (2014) J Biol Chem. , vol.289 , pp. 33568-33589
    • Gutierrez, E.1    Richardson, D.R.2    Jansson, P.J.3
  • 309
    • 84898065350 scopus 로고    scopus 로고
    • The metastasis suppressor, N-myc downstream-regulated gene 1 (NDRG1), inhibits stress-induced autophagy in cancer cells
    • Sahni S, Bae DH, Lane DJ, Kovacevic Z, Kalinowski DS, Jansson PJ, Richardson DR. The metastasis suppressor, N-myc downstream-regulated gene 1 (NDRG1), inhibits stress-induced autophagy in cancer cells. J Biol Chem. 2014; 289:9692-9709.
    • (2014) J Biol Chem. , vol.289 , pp. 9692-9709
    • Sahni, S.1    Bae, D.H.2    Lane, D.J.3    Kovacevic, Z.4    Kalinowski, D.S.5    Jansson, P.J.6    Richardson, D.R.7
  • 312
    • 78349308821 scopus 로고    scopus 로고
    • Negative regulation of autophagy
    • Liang C. Negative regulation of autophagy. Cell Death Differ. 2010; 17:1807-1815.
    • (2010) Cell Death Differ. , vol.17 , pp. 1807-1815
    • Liang, C.1
  • 313
    • 84921668375 scopus 로고    scopus 로고
    • Deferoxamine inhibits TRAIL-mediated apoptosis via regulation of autophagy in human colon cancer cells
    • Moon JH, Jeong JK, Park SY. Deferoxamine inhibits TRAIL-mediated apoptosis via regulation of autophagy in human colon cancer cells. Oncol Rep. 2015; 33:1171-1176.
    • (2015) Oncol Rep. , vol.33 , pp. 1171-1176
    • Moon, J.H.1    Jeong, J.K.2    Park, S.Y.3
  • 314
    • 0023281053 scopus 로고
    • Intracellular fate of ferritin in HeLa cells following microinjection
    • Hultcrantz R, Glaumann H. Intracellular fate of ferritin in HeLa cells following microinjection. Exp Cell Res. 1987; 171:203-212.
    • (1987) Exp Cell Res. , vol.171 , pp. 203-212
    • Hultcrantz, R.1    Glaumann, H.2
  • 315
  • 316
    • 73349099034 scopus 로고    scopus 로고
    • Specific iron chelators determine the route of ferritin degradation
    • De Domenico I, Ward DM, Kaplan J. Specific iron chelators determine the route of ferritin degradation. Blood. 2009; 114:4546-4551.
    • (2009) Blood. , vol.114 , pp. 4546-4551
    • De Domenico, I.1    Ward, D.M.2    Kaplan, J.3
  • 317
    • 75749122303 scopus 로고    scopus 로고
    • Methods in mammalian autophagy research
    • Mizushima N, Yoshimori T, Levine B. Methods in mammalian autophagy research. Cell. 2010; 140:313-326.
    • (2010) Cell. , vol.140 , pp. 313-326
    • Mizushima, N.1    Yoshimori, T.2    Levine, B.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.