메뉴 건너뛰기




Volumn 12, Issue 9, 2011, Pages 1322-1350

The complex biology of FOXO

Author keywords

Cancer; Cell cycle; Cell differentiation; Coactivator; FOXO; Insulin signaling; Metabolism; Transcription factor

Indexed keywords

ATROGIN 1; CCAAT ENHANCER BINDING PROTEIN BETA; CYCLIC AMP RESPONSIVE ELEMENT BINDING PROTEIN; DEXAMETHASONE; GLYCOGEN SYNTHASE KINASE 3ALPHA; GLYCOGEN SYNTHASE KINASE 3BETA; HEPATOCYTE NUCLEAR FACTOR 4; MANGANESE SUPEROXIDE DISMUTASE; MYOCARDIN; NOTCH RECEPTOR; PEROXISOME PROLIFERATOR ACTIVATED RECEPTOR GAMMA 2; PHOSPHATIDYLINOSITOL 3 KINASE; PHOSPHOENOLPYRUVATE CARBOXYKINASE (PYROPHOSPHATE); PROTEIN P300; PYRUVATE DEHYDROGENASE KINASE 4; SELENOPROTEIN P; SIRTUIN 1; SIRTUIN 2; SIRTUIN 3; SMAD PROTEIN; SOMATOMEDIN B; TRANSCRIPTION FACTOR FKHR; TRANSCRIPTION FACTOR FKHRL1; TRANSCRIPTION FACTOR FOXO; TRANSCRIPTION FACTOR FOXO4; TRANSCRIPTION FACTOR FOXO6; TRANSCRIPTION FACTOR SP1; UNCLASSIFIED DRUG; UNINDEXED DRUG; VASCULOTROPIN; WNT PROTEIN;

EID: 79959961108     PISSN: 13894501     EISSN: 18735592     Source Type: Journal    
DOI: 10.2174/138945011796150307     Document Type: Review
Times cited : (105)

References (440)
  • 1
    • 0024973841 scopus 로고
    • The homeotic gene fork head encodes a nuclear protein and is expressed in the terminal regions of the Drosophila embryo
    • Weigel D, Jurgens G, Kuttner F, Seifert E, Jackle H. The homeotic gene fork head encodes a nuclear protein and is expressed in the terminal regions of the Drosophila embryo. Cell 1989; 57: 645-658.
    • (1989) Cell , vol.57 , pp. 645-658
    • Weigel, D.1    Jurgens, G.2    Kuttner, F.3    Seifert, E.4    Jackle, H.5
  • 2
    • 62549108593 scopus 로고    scopus 로고
    • The evolution of Fox genes and their role in development and disease
    • Hannenhalli S, Kaestner KH. The evolution of Fox genes and their role in development and disease. Nat Rev Genet 2009; 10: 233-240.
    • (2009) Nat Rev Genet , vol.10 , pp. 233-240
    • Hannenhalli, S.1    Kaestner, K.H.2
  • 3
    • 0027959091 scopus 로고
    • Cloning and characterization of the t(X;11) breakpoint from a leukemic cell line identify a new member of the forkhead gene family
    • Parry P, Wei Y, Evans G. Cloning and characterization of the t(X;11) breakpoint from a leukemic cell line identify a new member of the forkhead gene family. Genes Chromosomes Cancer 1994; 11: 79-84.
    • (1994) Genes Chromosomes Cancer , vol.11 , pp. 79-84
    • Parry, P.1    Wei, Y.2    Evans, G.3
  • 4
    • 0031031378 scopus 로고    scopus 로고
    • Cloning and characterization of AFX, the gene that fuses to MLL in acute leukemias with a t(X;11)(q13;q23)
    • Borkhardt A, Repp R, Haas OA, et al. Cloning and characterization of AFX, the gene that fuses to MLL in acute leukemias with a t(X;11)(q13;q23). Oncogene 1997; 14: 195-202.
    • (1997) Oncogene , vol.14 , pp. 195-202
    • Borkhardt, A.1    Repp, R.2    Haas, O.A.3
  • 5
    • 0032518255 scopus 로고    scopus 로고
    • Cloning and characterization of three human forkhead genes that comprise an FKHR-like gene subfamily
    • Anderson MJ, Viars CS, Czekay S, Cavenee WK, Arden KC. Cloning and characterization of three human forkhead genes that comprise an FKHR-like gene subfamily. Genomics 1998; 47: 187-199.
    • (1998) Genomics , vol.47 , pp. 187-199
    • Anderson, M.J.1    Viars, C.S.2    Czekay, S.3    Cavenee, W.K.4    Arden, K.C.5
  • 7
    • 11144294668 scopus 로고    scopus 로고
    • From worm to human: Bioinformatics approaches to identify FOXO target genes
    • Xuan Z, Zhang MQ. From worm to human: bioinformatics approaches to identify FOXO target genes. Mech Ageing Dev 2005; 126: 209-215.
    • (2005) Mech Ageing Dev , vol.126 , pp. 209-215
    • Xuan, Z.1    Zhang, M.Q.2
  • 8
    • 0034661826 scopus 로고    scopus 로고
    • Identification of the differential distribution patterns of mRNAs and consensus binding sequences for mouse DAF-16 homologues
    • Furuyama T, Nakazawa T, Nakano I, Mori N. Identification of the differential distribution patterns of mRNAs and consensus binding sequences for mouse DAF-16 homologues. Biochem J 2000; 349: 629-634.
    • (2000) Biochem J , vol.349 , pp. 629-634
    • Furuyama, T.1    Nakazawa, T.2    Nakano, I.3    Mori, N.4
  • 9
    • 0036893049 scopus 로고    scopus 로고
    • Solution structure of the DNA-binding domain of interleukin enhancer binding factor 1 (FOXK1a)
    • Liu PP, Chen YC, Li C, et al. Solution structure of the DNA-binding domain of interleukin enhancer binding factor 1 (FOXK1a). Proteins 2002; 49: 543-553.
    • (2002) Proteins , vol.49 , pp. 543-553
    • Liu, P.P.1    Chen, Y.C.2    Li, C.3
  • 10
    • 0035918516 scopus 로고    scopus 로고
    • Solution structure of the DNA binding domain of the human forkhead transcription factor AFX (FOXO4)
    • Weigelt J, Climent I, Dahlman-Wright K, Wikstrom M. Solution structure of the DNA binding domain of the human forkhead transcription factor AFX (FOXO4). Biochemistry 2001; 40: 5861-5869.
    • (2001) Biochemistry , vol.40 , pp. 5861-5869
    • Weigelt, J.1    Climent, I.2    Dahlman-Wright, K.3    Wikstrom, M.4
  • 11
    • 36749079110 scopus 로고    scopus 로고
    • Crystal structure of the human FOXO3a-DBD/DNA complex suggests the effects of post-translational modification
    • Tsai KL, Sun YJ, Huang CY, Yang JY, Hung MC, Hsiao CD. Crystal structure of the human FOXO3a-DBD/DNA complex suggests the effects of post-translational modification. Nucleic Acids Res 2007; 35: 6984-6994.
    • (2007) Nucleic Acids Res , vol.35 , pp. 6984-6994
    • Tsai, K.L.1    Sun, Y.J.2    Huang, C.Y.3    Yang, J.Y.4    Hung, M.C.5    Hsiao, C.D.6
  • 12
    • 50849130900 scopus 로고    scopus 로고
    • Structural basis for DNA recognition by FoxO1 and its regulation by posttranslational modification
    • Brent MM, Anand R, Marmorstein R. Structural basis for DNA recognition by FoxO1 and its regulation by posttranslational modification. Structure 2008; 16: 1407-1416.
    • (2008) Structure , vol.16 , pp. 1407-1416
    • Brent, M.M.1    Anand, R.2    Marmorstein, R.3
  • 13
    • 0027270989 scopus 로고
    • Co-crystal structure of the HNF-3/fork head DNA-recognition motif resembles histone H5
    • Clark KL, Halay ED, Lai E, Burley SK. Co-crystal structure of the HNF-3/fork head DNA-recognition motif resembles histone H5. Nature 1993; 364: 412-420.
    • (1993) Nature , vol.364 , pp. 412-420
    • Clark, K.L.1    Halay, E.D.2    Lai, E.3    Burley, S.K.4
  • 14
    • 37249054901 scopus 로고    scopus 로고
    • Chromatin opening and stable perturbation of core histone: DNA contacts by FoxO1
    • Hatta M, Cirillo LA. Chromatin opening and stable perturbation of core histone: DNA contacts by FoxO1. J Biol Chem 2007; 282: 35583-35593.
    • (2007) J Biol Chem , vol.282 , pp. 35583-35593
    • Hatta, M.1    Cirillo, L.A.2
  • 15
    • 0027402969 scopus 로고    scopus 로고
    • Crystal structure of globular domain of histone H5 and its implications for nucleosome binding
    • 1993 Mar 18
    • Ramakrishnan V, Finch JT, Graziano V, Lee PL, Sweet RM. Crystal structure of globular domain of histone H5 and its implications for nucleosome binding. Nature. 1993 Mar 18;362(6417):219-223.
    • Nature , vol.362 , Issue.6417 , pp. 219-223
    • Ramakrishnan, V.1    Finch, J.T.2    Graziano, V.3    Lee, P.L.4    Sweet, R.M.5
  • 16
    • 0033388099 scopus 로고    scopus 로고
    • An early developmental transcription factor complex that is more stable on nucleosome core particles than on free DNA
    • Cirillo LA, Zaret KS. An early developmental transcription factor complex that is more stable on nucleosome core particles than on free DNA. Mol Cell 1999; 4: 961-969.
    • (1999) Mol Cell , vol.4 , pp. 961-969
    • Cirillo, L.A.1    Zaret, K.S.2
  • 17
    • 33645227508 scopus 로고    scopus 로고
    • The forkhead transcription factor FoxI1 remains bound to condensed mitotic chromosomes and stably remodels chromatin structure
    • Yan J, Xu L, Crawford G, Wang Z, Burgess SM. The forkhead transcription factor FoxI1 remains bound to condensed mitotic chromosomes and stably remodels chromatin structure. Mol Cell Biol 2006; 26: 155-168.
    • (2006) Mol Cell Biol , vol.26 , pp. 155-168
    • Yan, J.1    Xu, L.2    Crawford, G.3    Wang, Z.4    Burgess, S.M.5
  • 18
    • 0036184236 scopus 로고    scopus 로고
    • Opening of compacted chromatin by early developmental transcription factors HNF3 (FoxA) and GATA-4
    • Cirillo LA, Lin FR, Cuesta I, Friedman D, Jarnik M, Zaret KS. Opening of compacted chromatin by early developmental transcription factors HNF3 (FoxA) and GATA-4. Mol Cell 2002; 9: 279-289.
    • (2002) Mol Cell , vol.9 , pp. 279-289
    • Cirillo, L.A.1    Lin, F.R.2    Cuesta, I.3    Friedman, D.4    Jarnik, M.5    Zaret, K.S.6
  • 20
    • 0037438589 scopus 로고    scopus 로고
    • Common mechanism for oncogenic activation of MLL by forkhead family proteins
    • So CW, Cleary ML. Common mechanism for oncogenic activation of MLL by forkhead family proteins. Blood 2003; 101: 633-639.
    • (2003) Blood , vol.101 , pp. 633-639
    • So, C.W.1    Cleary, M.L.2
  • 21
    • 0036723650 scopus 로고    scopus 로고
    • MLL-AFX requires the transcriptional effector domains of AFX to transform myeloid progenitors and transdominantly interfere with forkhead protein function
    • So CW, Cleary ML. MLL-AFX requires the transcriptional effector domains of AFX to transform myeloid progenitors and transdominantly interfere with forkhead protein function. Mol Cell Biol 2002; 22: 6542-6552.
    • (2002) Mol Cell Biol , vol.22 , pp. 6542-6552
    • So, C.W.1    Cleary, M.L.2
  • 22
    • 34247267098 scopus 로고    scopus 로고
    • Both the N-terminal loop and wing W2 of the forkhead domain of transcription factor Foxo4 are important for DNA binding
    • Boura E, Silhan J, Herman P, et al. Both the N-terminal loop and wing W2 of the forkhead domain of transcription factor Foxo4 are important for DNA binding. J Biol Chem 2007; 282: 8265-8275.
    • (2007) J Biol Chem , vol.282 , pp. 8265-8275
    • Boura, E.1    Silhan, J.2    Herman, P.3
  • 23
    • 84954358441 scopus 로고    scopus 로고
    • Biochemical and structural characterization of an intramolecular interaction in FOXO3a and its binding with p53
    • Wang F, Marshall CB, Yamamoto K, et al. Biochemical and structural characterization of an intramolecular interaction in FOXO3a and its binding with p53. J Mol Biol 2008; 384: 590-603.
    • (2008) J Mol Biol , vol.384 , pp. 590-603
    • Wang, F.1    Marshall, C.B.2    Yamamoto, K.3
  • 24
    • 73649087930 scopus 로고    scopus 로고
    • Synergistic Interplay between Promoter Recognition and CBP/p300 Coactivator Recruitment by FOXO3a
    • Wang F, Marshall CB, Li GY, Yamamoto K, Mak TW, Ikura M. Synergistic Interplay between Promoter Recognition and CBP/p300 Coactivator Recruitment by FOXO3a. ACS Chem Biol 2009; 4: 1017-1027.
    • (2009) ACS Chem Biol , vol.4 , pp. 1017-1027
    • Wang, F.1    Marshall, C.B.2    Li, G.Y.3    Yamamoto, K.4    Mak, T.W.5    Ikura, M.6
  • 26
    • 0038003956 scopus 로고    scopus 로고
    • Decisions on life and death: FOXO Forkhead transcription factors are in command when PKB/Akt is off duty
    • Burgering BM, Medema RH. Decisions on life and death: FOXO Forkhead transcription factors are in command when PKB/Akt is off duty. J Leukoc Biol 2003; 73: 689-701.
    • (2003) J Leukoc Biol , vol.73 , pp. 689-701
    • Burgering, B.M.1    Medema, R.H.2
  • 27
    • 1542267804 scopus 로고    scopus 로고
    • Disruption of forkhead transcription factor (FOXO) family members in mice reveals their functional diversification
    • Hosaka T, Biggs WH, 3rd, Tieu D, et al. Disruption of forkhead transcription factor (FOXO) family members in mice reveals their functional diversification. Proc Natl Acad Sci USA 2004; 101: 2975-2980.
    • (2004) Proc Natl Acad Sci USA , vol.101 , pp. 2975-2980
    • Hosaka, T.1    Biggs, W.H.2    Tieu, D.3
  • 28
    • 4544227785 scopus 로고    scopus 로고
    • Abnormal angiogenesis in Foxo1 (Fkhr)-deficient mice
    • Furuyama T, Kitayama K, Shimoda Y, et al. Abnormal angiogenesis in Foxo1 (Fkhr)-deficient mice. J Biol Chem 2004; 279: 34741-34749.
    • (2004) J Biol Chem , vol.279 , pp. 34741-34749
    • Furuyama, T.1    Kitayama, K.2    Shimoda, Y.3
  • 29
    • 0038152845 scopus 로고    scopus 로고
    • Suppression of ovarian follicle activation in mice by the transcription factor Foxo3a
    • Castrillon DH, Miao L, Kollipara R, Horner JW, DePinho RA. Suppression of ovarian follicle activation in mice by the transcription factor Foxo3a. Science 2003; 301: 215-218.
    • (2003) Science , vol.301 , pp. 215-218
    • Castrillon, D.H.1    Miao, L.2    Kollipara, R.3    Horner, J.W.4    Depinho, R.A.5
  • 30
    • 4143129696 scopus 로고    scopus 로고
    • Regulation of NF-kappaB, Th activation, and autoinflammation by the forkhead transcription factor Foxo3a
    • Lin L, Hron JD, Peng SL. Regulation of NF-kappaB, Th activation, and autoinflammation by the forkhead transcription factor Foxo3a. Immunity 2004; 21: 203-213.
    • (2004) Immunity , vol.21 , pp. 203-213
    • Lin, L.1    Hron, J.D.2    Peng, S.L.3
  • 31
    • 0036651790 scopus 로고    scopus 로고
    • A novel mechanism of gene regulation and tumor suppression by the transcription factor FKHR
    • Ramaswamy S, Nakamura N, Sansal I, Bergeron L, Sellers WR. A novel mechanism of gene regulation and tumor suppression by the transcription factor FKHR. Cancer Cell 2002; 2: 81-91.
    • (2002) Cancer Cell , vol.2 , pp. 81-91
    • Ramaswamy, S.1    Nakamura, N.2    Sansal, I.3    Bergeron, L.4    Sellers, W.R.5
  • 32
    • 0033595011 scopus 로고    scopus 로고
    • Protein kinase B/Akt-mediated phosphorylation promotes nuclear exclusion of the winged helix transcription factor FKHR1
    • Biggs WH, 3rd, Meisenhelder J, Hunter T, Cavenee WK, Arden KC. Protein kinase B/Akt-mediated phosphorylation promotes nuclear exclusion of the winged helix transcription factor FKHR1. Proc Natl Acad Sci USA 1999; 96: 7421-7426.
    • (1999) Proc Natl Acad Sci USA , vol.96 , pp. 7421-7426
    • Biggs, W.H.1    Meisenhelder, J.2    Hunter, T.3    Cavenee, W.K.4    Arden, K.C.5
  • 33
    • 30444442827 scopus 로고    scopus 로고
    • Spatial and temporal expression of FoxO transcription factors in the developing and adult murine brain
    • Hoekman MF, Jacobs FM, Smidt MP, Burbach JP. Spatial and temporal expression of FoxO transcription factors in the developing and adult murine brain. Gene Expr Patterns 2006; 6: 134-140.
    • (2006) Gene Expr Patterns , vol.6 , pp. 134-140
    • Hoekman, M.F.1    Jacobs, F.M.2    Smidt, M.P.3    Burbach, J.P.4
  • 34
    • 36448968532 scopus 로고    scopus 로고
    • FoxO3 coordinately activates protein degradation by the autophagic/lysosomal and proteasomal pathways in atrophying muscle cells
    • Zhao J, Brault JJ, Schild A, et al. FoxO3 coordinately activates protein degradation by the autophagic/lysosomal and proteasomal pathways in atrophying muscle cells. Cell Metab 2007; 6: 472-483.
    • (2007) Cell Metab , vol.6 , pp. 472-483
    • Zhao, J.1    Brault, J.J.2    Schild, A.3
  • 35
    • 0042092531 scopus 로고    scopus 로고
    • Genes that act downstream of DAF-16 to influence the lifespan of Caenorhabditis elegans
    • Murphy CT, McCarroll SA, Bargmann CI, et al. Genes that act downstream of DAF-16 to influence the lifespan of Caenorhabditis elegans. Nature 2003; 424: 277-283.
    • (2003) Nature , vol.424 , pp. 277-283
    • Murphy, C.T.1    McCarroll, S.A.2    Bargmann, C.I.3
  • 36
    • 36448940798 scopus 로고    scopus 로고
    • FoxO3 controls autophagy in skeletal muscle in vivo
    • Mammucari C, Milan G, Romanello V, et al. FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab 2007; 6: 458-471.
    • (2007) Cell Metab , vol.6 , pp. 458-471
    • Mammucari, C.1    Milan, G.2    Romanello, V.3
  • 37
    • 0037466652 scopus 로고    scopus 로고
    • DAF-16 target genes that control C. elegans life-span and metabolism
    • Lee SS, Kennedy S, Tolonen AC, Ruvkun G. DAF-16 target genes that control C. elegans life-span and metabolism. Science 2003; 300: 644-647.
    • (2003) Science , vol.300 , pp. 644-647
    • Lee, S.S.1    Kennedy, S.2    Tolonen, A.C.3    Ruvkun, G.4
  • 38
    • 0037134040 scopus 로고    scopus 로고
    • DNA repair pathway stimulated by the forkhead transcription factor FOXO3a through the Gadd45 protein
    • Tran H, Brunet A, Grenier JM, et al. DNA repair pathway stimulated by the forkhead transcription factor FOXO3a through the Gadd45 protein. Science 2002; 296: 530-534.
    • (2002) Science , vol.296 , pp. 530-534
    • Tran, H.1    Brunet, A.2    Grenier, J.M.3
  • 39
    • 0037192473 scopus 로고    scopus 로고
    • Redox regulation of forkhead proteins through a p66shc-dependent signaling pathway
    • Nemoto S, Finkel T. Redox regulation of forkhead proteins through a p66shc-dependent signaling pathway. Science 2002; 295: 2450-2452.
    • (2002) Science , vol.295 , pp. 2450-2452
    • Nemoto, S.1    Finkel, T.2
  • 40
    • 0034643331 scopus 로고    scopus 로고
    • AFX-like Forkhead transcription factors mediate cell-cycle regulation by Ras and PKB through p27kip1
    • Medema RH, Kops GJ, Bos JL, Burgering BM. AFX-like Forkhead transcription factors mediate cell-cycle regulation by Ras and PKB through p27kip1. Nature 2000; 404: 782-787.
    • (2000) Nature , vol.404 , pp. 782-787
    • Medema, R.H.1    Kops, G.J.2    Bos, J.L.3    Burgering, B.M.4
  • 41
    • 0037136563 scopus 로고    scopus 로고
    • Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress
    • Kops GJ, Dansen TB, Polderman PE, et al. Forkhead transcription factor FOXO3a protects quiescent cells from oxidative stress. Nature 2002; 419: 316-321.
    • (2002) Nature , vol.419 , pp. 316-321
    • Kops, G.J.1    Dansen, T.B.2    Polderman, P.E.3
  • 42
    • 0034609737 scopus 로고    scopus 로고
    • Expression of the pro-apoptotic Bcl-2 family member Bim is regulated by the forkhead transcription factor FKHR-L1
    • Dijkers PF, Medema RH, Lammers JW, Koenderman L, Coffer PJ. Expression of the pro-apoptotic Bcl-2 family member Bim is regulated by the forkhead transcription factor FKHR-L1. Curr Biol 2000; 10: 1201-1214.
    • (2000) Curr Biol , vol.10 , pp. 1201-1214
    • Dijkers, P.F.1    Medema, R.H.2    Lammers, J.W.3    Koenderman, L.4    Coffer, P.J.5
  • 43
    • 0033582929 scopus 로고    scopus 로고
    • Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor
    • Brunet A, Bonni A, Zigmond MJ, et al. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 1999; 96: 857-868.
    • (1999) Cell , vol.96 , pp. 857-868
    • Brunet, A.1    Bonni, A.2    Zigmond, M.J.3
  • 44
    • 70350518100 scopus 로고    scopus 로고
    • Chemical interrogation of FOXO3a nuclear translocation identifies potent and selective inhibitors of phosphoinositide 3-kinases
    • Link W, Oyarzabal J, Serelde BG, et al. Chemical interrogation of FOXO3a nuclear translocation identifies potent and selective inhibitors of phosphoinositide 3-kinases. J Biol Chem 2009; 284: 28392-28400.
    • (2009) J Biol Chem , vol.284 , pp. 28392-28400
    • Link, W.1    Oyarzabal, J.2    Serelde, B.G.3
  • 45
    • 11144357466 scopus 로고    scopus 로고
    • IkappaB kinase promotes tumorigenesis through inhibition of forkhead FOXO3a
    • Hu MC, Lee DF, Xia W, et al. IkappaB kinase promotes tumorigenesis through inhibition of forkhead FOXO3a. Cell 2004; 117: 225-237.
    • (2004) Cell , vol.117 , pp. 225-237
    • Mc, H.1    Lee, D.F.2    Xia, W.3
  • 46
    • 16244415180 scopus 로고    scopus 로고
    • Constitutively active FOXO4 inhibits Akt activity, regulates p27 Kip1 stability, and suppresses HER2-mediated tumorigenicity
    • Yang H, Zhao R, Yang HY, Lee MH. Constitutively active FOXO4 inhibits Akt activity, regulates p27 Kip1 stability, and suppresses HER2-mediated tumorigenicity. Oncogene 2005; 24: 1924-1935.
    • (2005) Oncogene , vol.24 , pp. 1924-1935
    • Yang, H.1    Zhao, R.2    Yang, H.Y.3    Lee, M.H.4
  • 47
    • 12144290563 scopus 로고    scopus 로고
    • Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase
    • Brunet A, Sweeney LB, Sturgill JF, et al. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 2004; 303: 2011-2015.
    • (2004) Science , vol.303 , pp. 2011-2015
    • Brunet, A.1    Sweeney, L.B.2    Sturgill, J.F.3
  • 48
    • 1642332084 scopus 로고    scopus 로고
    • Integration of Smad and forkhead pathways in the control of neuroepithelial and glioblastoma cell proliferation
    • Seoane J, Le HV, Shen L, Anderson SA, Massague J. Integration of Smad and forkhead pathways in the control of neuroepithelial and glioblastoma cell proliferation. Cell 2004; 117: 211-223.
    • (2004) Cell , vol.117 , pp. 211-223
    • Seoane, J.1    Le, H.V.2    Shen, L.3    Anderson, S.A.4    Massague, J.5
  • 51
    • 4143050290 scopus 로고    scopus 로고
    • The interaction between FOXO and SIRT1: Tipping the balance towards survival
    • Giannakou ME, Partridge L. The interaction between FOXO and SIRT1: tipping the balance towards survival. Trends Cell Biol 2004; 14: 408-412.
    • (2004) Trends Cell Biol , vol.14 , pp. 408-412
    • Giannakou, M.E.1    Partridge, L.2
  • 52
    • 2942563805 scopus 로고    scopus 로고
    • Drosophila dFOXO controls lifespan and regulates insulin signalling in brain and fat body
    • Hwangbo DS, Gershman B, Tu MP, Palmer M, Tatar M. Drosophila dFOXO controls lifespan and regulates insulin signalling in brain and fat body. Nature 2004; 429: 562-566.
    • (2004) Nature , vol.429 , pp. 562-566
    • Hwangbo, D.S.1    Gershman, B.2    Tu, M.P.3    Palmer, M.4    Tatar, M.5
  • 53
    • 0035846604 scopus 로고    scopus 로고
    • Daf-16 integrates developmental and environmental inputs to mediate aging in the nematode Caenorhabditis elegans
    • Henderson ST, Johnson TE. daf-16 integrates developmental and environmental inputs to mediate aging in the nematode Caenorhabditis elegans. Curr Biol 2001; 11: 1975-1980.
    • (2001) Curr Biol , vol.11 , pp. 1975-1980
    • Henderson, S.T.1    Johnson, T.E.2
  • 54
    • 0030657540 scopus 로고    scopus 로고
    • Daf-16: An HNF-3/forkhead family member that can function to double the life-span of Caenorhabditis elegans
    • Lin K, Dorman JB, Rodan A, Kenyon C. daf-16: An HNF-3/forkhead family member that can function to double the life-span of Caenorhabditis elegans. Science 1997; 278: 1319-1322.
    • (1997) Science , vol.278 , pp. 1319-1322
    • Lin, K.1    Dorman, J.B.2    Rodan, A.3    Kenyon, C.4
  • 55
    • 0030659557 scopus 로고    scopus 로고
    • The Fork head transcription factor DAF-16 transduces insulin-like metabolic and longevity signals in C. elegans
    • Ogg S, Paradis S, Gottlieb S, et al. The Fork head transcription factor DAF-16 transduces insulin-like metabolic and longevity signals in C. elegans. Nature 1997; 389: 994-999.
    • (1997) Nature , vol.389 , pp. 994-999
    • Ogg, S.1    Paradis, S.2    Gottlieb, S.3
  • 56
    • 62449083712 scopus 로고    scopus 로고
    • Association of FOXO3A variation with human longevity confirmed in German centenarians
    • Flachsbart F, Caliebe A, Kleindorp R, et al. Association of FOXO3A variation with human longevity confirmed in German centenarians. Proc Natl Acad Sci USA 2009; 106: 2700-2705.
    • (2009) Proc Natl Acad Sci USA , vol.106 , pp. 2700-2705
    • Flachsbart, F.1    Caliebe, A.2    Kleindorp, R.3
  • 57
    • 52949122885 scopus 로고    scopus 로고
    • FOXO3A genotype is strongly associated with human longevity
    • Willcox BJ, Donlon TA, He Q, et al. FOXO3A genotype is strongly associated with human longevity. Proc Natl Acad Sci USA 2008; 105:13987-13992.
    • (2008) Proc Natl Acad Sci USA , vol.105 , pp. 13987-13992
    • Willcox, B.J.1    Donlon, T.A.2    He, Q.3
  • 58
    • 33846419112 scopus 로고    scopus 로고
    • FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress
    • Tothova Z, Kollipara R, Huntly BJ, et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell 2007; 128: 325-339.
    • (2007) Cell , vol.128 , pp. 325-339
    • Tothova, Z.1    Kollipara, R.2    Huntly, B.J.3
  • 59
    • 34249882777 scopus 로고    scopus 로고
    • Foxo3a is essential for maintenance of the hematopoietic stem cell pool
    • Miyamoto K, Araki KY, Naka K, et al. Foxo3a is essential for maintenance of the hematopoietic stem cell pool. Cell Stem Cell 2007; 1: 101-112.
    • (2007) Cell Stem Cell , vol.1 , pp. 101-112
    • Miyamoto, K.1    Araki, K.Y.2    Naka, K.3
  • 60
    • 1642580499 scopus 로고    scopus 로고
    • FoxO3a regulates erythroid differentiation and induces BTG1, an activator of protein arginine methyl transferase 1
    • Bakker WJ, Blazquez-Domingo M, Kolbus A, et al. FoxO3a regulates erythroid differentiation and induces BTG1, an activator of protein arginine methyl transferase 1. J Cell Biol 2004; 164: 175-184.
    • (2004) J Cell Biol , vol.164 , pp. 175-184
    • Bakker, W.J.1    Blazquez-Domingo, M.2    Kolbus, A.3
  • 61
    • 34250214921 scopus 로고    scopus 로고
    • Foxs and Ets in the transcriptional regulation of endothelial cell differentiation and angiogenesis
    • Dejana E, Taddei A, Randi AM. Foxs and Ets in the transcriptional regulation of endothelial cell differentiation and angiogenesis. Biochim Biophys Acta 2007; 1775: 298-312.
    • (2007) Biochim Biophys Acta , vol.1775 , pp. 298-312
    • Dejana, E.1    Taddei, A.2    Randi, A.M.3
  • 62
    • 0042477748 scopus 로고    scopus 로고
    • Regulation of insulin-like growth factor-dependent myoblast differentiation by Foxo forkhead transcription factors
    • Hribal ML, Nakae J, Kitamura T, Shutter JR, Accili D. Regulation of insulin-like growth factor-dependent myoblast differentiation by Foxo forkhead transcription factors. J Cell Biol 2003; 162: 535-541.
    • (2003) J Cell Biol , vol.162 , pp. 535-541
    • Hribal, M.L.1    Nakae, J.2    Kitamura, T.3    Shutter, J.R.4    Accili, D.5
  • 63
    • 0037237279 scopus 로고    scopus 로고
    • The forkhead transcription factor Foxo1 regulates adipocyte differentiation
    • Nakae J, Kitamura T, Kitamura Y, Biggs WH, 3rd, Arden KC, Accili D. The forkhead transcription factor Foxo1 regulates adipocyte differentiation. Dev Cell 2003; 4: 119-129.
    • (2003) Dev Cell , vol.4 , pp. 119-129
    • Nakae, J.1    Kitamura, T.2    Kitamura, Y.3    Biggs, W.H.4    Arden, K.C.5    Accili, D.6
  • 64
    • 34548349302 scopus 로고    scopus 로고
    • Impaired regulation of hepatic glucose production in mice lacking the forkhead transcription factor Foxo1 in liver
    • Matsumoto M, Pocai A, Rossetti L, Depinho RA, Accili D. Impaired regulation of hepatic glucose production in mice lacking the forkhead transcription factor Foxo1 in liver. Cell Metab 2007; 6: 208-216.
    • (2007) Cell Metab , vol.6 , pp. 208-216
    • Matsumoto, M.1    Pocai, A.2    Rossetti, L.3    Depinho, R.A.4    Accili, D.5
  • 65
    • 33748312093 scopus 로고    scopus 로고
    • Dual role of transcription factor FoxO1 in controlling hepatic insulin sensitivity and lipid metabolism
    • Matsumoto M, Han S, Kitamura T, Accili D. Dual role of transcription factor FoxO1 in controlling hepatic insulin sensitivity and lipid metabolism. J Clin Invest 2006; 116: 2464-2472.
    • (2006) J Clin Invest , vol.116 , pp. 2464-2472
    • Matsumoto, M.1    Han, S.2    Kitamura, T.3    Accili, D.4
  • 66
    • 33745576798 scopus 로고    scopus 로고
    • Role of hypothalamic Foxo1 in the regulation of food intake and energy homeostasis
    • Kim MS, Pak YK, Jang PG, et al. Role of hypothalamic Foxo1 in the regulation of food intake and energy homeostasis. Nat Neurosci 2006; 9: 901-906.
    • (2006) Nat Neurosci , vol.9 , pp. 901-906
    • Kim, M.S.1    Pak, Y.K.2    Jang, P.G.3
  • 67
    • 0038187621 scopus 로고    scopus 로고
    • Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1alpha interaction
    • Puigserver P, Rhee J, Donovan J, et al. Insulin-regulated hepatic gluconeogenesis through FOXO1-PGC-1alpha interaction. Nature 2003; 423: 550-555.
    • (2003) Nature , vol.423 , pp. 550-555
    • Puigserver, P.1    Rhee, J.2    Donovan, J.3
  • 68
    • 33646590947 scopus 로고    scopus 로고
    • Forkhead protein FoxO1 mediates Agrp-dependent effects of leptin on food intake
    • Kitamura T, Feng Y, Kitamura YI, et al. Forkhead protein FoxO1 mediates Agrp-dependent effects of leptin on food intake. Nat Med 2006; 12: 534-540.
    • (2006) Nat Med , vol.12 , pp. 534-540
    • Kitamura, T.1    Feng, Y.2    Kitamura, Y.I.3
  • 69
    • 41849128523 scopus 로고    scopus 로고
    • The FoxO code
    • Calnan DR, Brunet A. The FoxO code. Oncogene 2008; 27: 2276-2288.
    • (2008) Oncogene , vol.27 , pp. 2276-2288
    • Calnan, D.R.1    Brunet, A.2
  • 70
    • 0034680839 scopus 로고    scopus 로고
    • Regulation of glucose-6-phosphatase gene expression by protein kinase Balpha and the forkhead transcription factor FKHR. Evidence for insulin response unit-dependent and -independent effects of insulin on promoter activity
    • Schmoll D, Walker KS, Alessi DR, et al. Regulation of glucose-6-phosphatase gene expression by protein kinase Balpha and the forkhead transcription factor FKHR. Evidence for insulin response unit-dependent and -independent effects of insulin on promoter activity. J Biol Chem 2000; 275: 36324-36333.
    • (2000) J Biol Chem , vol.275 , pp. 36324-36333
    • Schmoll, D.1    Walker, K.S.2    Alessi, D.R.3
  • 71
    • 0036787607 scopus 로고    scopus 로고
    • Regulation of insulin action and pancreatic beta-cell function by mutated alleles of the gene encoding forkhead transcription factor Foxo1
    • Nakae J, Biggs WH, 3rd, Kitamura T, et al. Regulation of insulin action and pancreatic beta-cell function by mutated alleles of the gene encoding forkhead transcription factor Foxo1. Nat Genet. 2002; 32: 245-253.
    • (2002) Nat Genet , vol.32 , pp. 245-253
    • Nakae, J.1    Biggs, W.H.2    Kitamura, T.3
  • 72
    • 0034730660 scopus 로고    scopus 로고
    • Regulation of phosphoenolpyruvate carboxykinase and insulin-like growth factor-binding protein-1 gene expression by insulin. The role of winged helix/forkhead proteins
    • Hall RK, Yamasaki T, Kucera T, Waltner-Law M, O'Brien R, Granner DK. Regulation of phosphoenolpyruvate carboxykinase and insulin-like growth factor-binding protein-1 gene expression by insulin. The role of winged helix/forkhead proteins. J Biol Chem 2000; 275: 30169-30175.
    • (2000) J Biol Chem , vol.275 , pp. 30169-30175
    • Hall, R.K.1    Yamasaki, T.2    Kucera, T.3    Waltner-Law, M.4    O'Brien, R.5    Granner, D.K.6
  • 73
    • 38049148308 scopus 로고    scopus 로고
    • FoxO transcription factors activate Akt and attenuate insulin signaling in heart by inhibiting protein phosphatases
    • Ni YG, Wang N, Cao DJ, et al. FoxO transcription factors activate Akt and attenuate insulin signaling in heart by inhibiting protein phosphatases. Proc Natl Acad Sci USA 2007; 104: 20517-20522.
    • (2007) Proc Natl Acad Sci USA , vol.104 , pp. 20517-20522
    • Ni, Y.G.1    Wang, N.2    Cao, D.J.3
  • 74
    • 34848850156 scopus 로고    scopus 로고
    • An AMPK-FOXO pathway mediates longevity induced by a novel method of dietary restriction in C. elegans
    • Greer EL, Dowlatshahi D, Banko MR, et al. An AMPK-FOXO pathway mediates longevity induced by a novel method of dietary restriction in C. elegans. Curr Biol 2007; 17: 1646-1656.
    • (2007) Curr Biol , vol.17 , pp. 1646-1656
    • Greer, E.L.1    Dowlatshahi, D.2    Banko, M.R.3
  • 75
    • 0030813398 scopus 로고    scopus 로고
    • Ruvkun G. daf-2, an insulin receptor-like gene that regulates longevity and diapause in Caenorhabditis elegans
    • Kimura KD, Tissenbaum HA, Liu Y, Ruvkun G. daf-2, an insulin receptor-like gene that regulates longevity and diapause in Caenorhabditis elegans. Science 1997; 277: 942-946.
    • (1997) Science , vol.277 , pp. 942-946
    • Kimura, K.D.1    Tissenbaum, H.A.2    Liu, Y.3
  • 76
    • 0027771804 scopus 로고
    • A C. elegans mutant that lives twice as long as wild type
    • Kenyon C, Chang J, Gensch E, Rudner A, Tabtiang R. A C. elegans mutant that lives twice as long as wild type. Nature 1993; 366: 461-464.
    • (1993) Nature , vol.366 , pp. 461-464
    • Kenyon, C.1    Chang, J.2    Gensch, E.3    Rudner, A.4    Tabtiang, R.5
  • 77
    • 0035958853 scopus 로고    scopus 로고
    • Forkhead homologue in rhabdomyosarcoma functions as a bifunctional nuclear receptor-interacting protein with both coactivator and corepressor functions
    • Zhao HH, Herrera RE, Coronado-Heinsohn E, et al. Forkhead homologue in rhabdomyosarcoma functions as a bifunctional nuclear receptor-interacting protein with both coactivator and corepressor functions. J Biol Chem 2001; 276: 27907-27912.
    • (2001) J Biol Chem , vol.276 , pp. 27907-27912
    • Zhao, H.H.1    Herrera, R.E.2    Coronado-Heinsohn, E.3
  • 78
    • 0038645860 scopus 로고    scopus 로고
    • Hepatocyte nuclear factor-4 is a novel downstream target of insulin via FKHR as a signal-regulated transcriptional inhibitor
    • Hirota K, Daitoku H, Matsuzaki H, et al. Hepatocyte nuclear factor-4 is a novel downstream target of insulin via FKHR as a signal-regulated transcriptional inhibitor. J Biol Chem 2003; 278: 13056-13060.
    • (2003) J Biol Chem , vol.278 , pp. 13056-13060
    • Hirota, K.1    Daitoku, H.2    Matsuzaki, H.3
  • 79
    • 0030695445 scopus 로고    scopus 로고
    • The maturity-onset diabetes of the young (MODY1) transcription factor HNF4alpha regulates expression of genes required for glucose transport and metabolism
    • Stoffel M, Duncan SA. The maturity-onset diabetes of the young (MODY1) transcription factor HNF4alpha regulates expression of genes required for glucose transport and metabolism. Proc Natl Acad Sci USA 1997; 94: 13209-13214.
    • (1997) Proc Natl Acad Sci USA , vol.94 , pp. 13209-13214
    • Stoffel, M.1    Duncan, S.A.2
  • 80
    • 0025690311 scopus 로고
    • Liver-enriched transcription factor HNF-4 is a novel member of the steroid hormone receptor superfamily
    • Sladek FM, Zhong WM, Lai E, Darnell JE, Jr. Liver-enriched transcription factor HNF-4 is a novel member of the steroid hormone receptor superfamily. Genes Dev 1990; 4: 2353-2365.
    • (1990) Genes Dev , vol.4 , pp. 2353-2365
    • Sladek, F.M.1    Zhong, W.M.2    Lai, E.3    Darnell Jr., J.E.4
  • 81
    • 0035141324 scopus 로고    scopus 로고
    • Hepatocyte nuclear factor 4alpha (nuclear receptor 2A1) is essential for maintenance of hepatic gene expression and lipid homeostasis
    • Hayhurst GP, Lee YH, Lambert G, Ward JM, Gonzalez FJ. Hepatocyte nuclear factor 4alpha (nuclear receptor 2A1) is essential for maintenance of hepatic gene expression and lipid homeostasis. Mol Cell Biol 2001; 21: 1393-1403.
    • (2001) Mol Cell Biol , vol.21 , pp. 1393-1403
    • Hayhurst, G.P.1    Lee, Y.H.2    Lambert, G.3    Ward, J.M.4    Gonzalez, F.J.5
  • 82
    • 0030042979 scopus 로고    scopus 로고
    • Human hepatocyte nuclear factor 4 isoforms are encoded by distinct and differentially expressed genes
    • Drewes T, Senkel S, Holewa B, Ryffel GU. Human hepatocyte nuclear factor 4 isoforms are encoded by distinct and differentially expressed genes. Mol Cell Biol 1996; 16: 925-931.
    • (1996) Mol Cell Biol , vol.16 , pp. 925-931
    • Drewes, T.1    Senkel, S.2    Holewa, B.3    Ryffel, G.U.4
  • 83
    • 0027362622 scopus 로고
    • Functional characterization of the L-type pyruvate kinase gene glucose response complex
    • Diaz Guerra MJ, Bergot MO, Martinez A, Cuif MH, Kahn A, Raymondjean M. Functional characterization of the L-type pyruvate kinase gene glucose response complex. Mol Cell Biol 1993; 13: 7725-7733.
    • (1993) Mol Cell Biol , vol.13 , pp. 7725-7733
    • Diaz, G.M.J.1    Bergot, M.O.2    Martinez, A.3    Cuif, M.H.4    Kahn, A.5    Raymondjean, M.6
  • 84
    • 57749116021 scopus 로고    scopus 로고
    • A combination of HNF-4 and Foxo1 is required for reciprocal transcriptional regulation of glucokinase and glucose-6-phosphatase genes in response to fasting and feeding
    • Hirota K, Sakamaki J, Ishida J, et al. A combination of HNF-4 and Foxo1 is required for reciprocal transcriptional regulation of glucokinase and glucose-6-phosphatase genes in response to fasting and feeding. J Biol Chem 2008; 283: 32432-32441.
    • (2008) J Biol Chem , vol.283 , pp. 32432-32441
    • Hirota, K.1    Sakamaki, J.2    Ishida, J.3
  • 85
    • 0038645860 scopus 로고    scopus 로고
    • Hepatocyte nuclear factor-4 is a novel downstream target of insulin via FKHR as a signal-regulated transcriptional inhibitor
    • Hirota K, Daitoku H, Matsuzaki H, et al. Hepatocyte nuclear factor-4 is a novel downstream target of insulin via FKHR as a signal-regulated transcriptional inhibitor. J Biol Chem 2003; 278: 13056-13060.
    • (2003) J Biol Chem , vol.278 , pp. 13056-13060
    • Hirota, K.1    Daitoku, H.2    Matsuzaki, H.3
  • 86
    • 0042232315 scopus 로고    scopus 로고
    • PGC-1beta in the regulation of hepatic glucose and energy metabolism
    • Lin J, Tarr PT, Yang R, et al. PGC-1beta in the regulation of hepatic glucose and energy metabolism. J Biol Chem 2003; 278: 30843-30848.
    • (2003) J Biol Chem , vol.278 , pp. 30843-30848
    • Lin, J.1    Tarr, P.T.2    Yang, R.3
  • 87
    • 0036903174 scopus 로고    scopus 로고
    • Adaptations of skeletal muscle to exercise: Rapid increase in the transcriptional coactivator PGC-1
    • Baar K, Wende AR, Jones TE, et al. Adaptations of skeletal muscle to exercise: rapid increase in the transcriptional coactivator PGC-1. Faseb J 2002; 16: 1879-1886.
    • (2002) Faseb J , vol.16 , pp. 1879-1886
    • Baar, K.1    Wende, A.R.2    Jones, T.E.3
  • 88
    • 0033538473 scopus 로고    scopus 로고
    • Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1
    • Wu Z, Puigserver P, Andersson U, et al. Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1. Cell 1999; 98: 115-124.
    • (1999) Cell , vol.98 , pp. 115-124
    • Wu, Z.1    Puigserver, P.2    Andersson, U.3
  • 89
    • 0032549811 scopus 로고    scopus 로고
    • A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis
    • Puigserver P, Wu Z, Park CW, Graves R, Wright M, Spiegelman BM. A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell 1998; 92: 829-839.
    • (1998) Cell , vol.92 , pp. 829-839
    • Puigserver, P.1    Wu, Z.2    Park, C.W.3    Graves, R.4    Wright, M.5    Spiegelman, B.M.6
  • 90
    • 5344252327 scopus 로고    scopus 로고
    • Defects in adaptive energy metabolism with CNS-linked hyperactivity in PGC-1alpha null mice
    • Lin J, Wu PH, Tarr PT, et al. Defects in adaptive energy metabolism with CNS-linked hyperactivity in PGC-1alpha null mice. Cell 2004; 119: 121-135.
    • (2004) Cell , vol.119 , pp. 121-135
    • Lin, J.1    Wu, P.H.2    Tarr, P.T.3
  • 91
    • 10744227070 scopus 로고    scopus 로고
    • Control of pancreas and liver gene expression by HNF transcription factors
    • Odom DT, Zizlsperger N, Gordon DB, et al. Control of pancreas and liver gene expression by HNF transcription factors. Science 2004; 303:1378-1381.
    • (2004) Science , vol.303 , pp. 1378-1381
    • Odom, D.T.1    Zizlsperger, N.2    Gordon, D.B.3
  • 92
    • 0242349197 scopus 로고    scopus 로고
    • Regulation of hepatic fasting response by PPARgamma coactivator-1alpha (PGC-1): Requirement for hepatocyte nuclear factor 4alpha in gluconeogenesis
    • Rhee J, Inoue Y, Yoon JC, et al. Regulation of hepatic fasting response by PPARgamma coactivator-1alpha (PGC-1): requirement for hepatocyte nuclear factor 4alpha in gluconeogenesis. Proc Natl Acad Sci USA 2003; 100: 4012-4017.
    • (2003) Proc Natl Acad Sci USA , vol.100 , pp. 4012-4017
    • Rhee, J.1    Inoue, Y.2    Yoon, J.C.3
  • 93
    • 0037342151 scopus 로고    scopus 로고
    • Regulation of PGC-1 promoter activity by protein kinase B and the forkhead transcription factor FKHR
    • Daitoku H, Yamagata K, Matsuzaki H, Hatta M, Fukamizu A. Regulation of PGC-1 promoter activity by protein kinase B and the forkhead transcription factor FKHR. Diabetes 2003; 52: 642-649.
    • (2003) Diabetes , vol.52 , pp. 642-649
    • Daitoku, H.1    Yamagata, K.2    Matsuzaki, H.3    Hatta, M.4    Fukamizu, A.5
  • 94
    • 64149111641 scopus 로고    scopus 로고
    • A PGC-1alpha-O-GlcNAc transferase complex regulates FoxO transcription factor activity in response to glucose
    • Housley MP, Udeshi ND, Rodgers JT, et al. A PGC-1alpha-O-GlcNAc transferase complex regulates FoxO transcription factor activity in response to glucose. J Biol Chem 2009; 284: 5148-5157.
    • (2009) J Biol Chem , vol.284 , pp. 5148-5157
    • Housley, M.P.1    Udeshi, N.D.2    Rodgers, J.T.3
  • 95
    • 34250740323 scopus 로고    scopus 로고
    • Akt/PKB regulates hepatic metabolism by directly inhibiting PGC-1alpha transcription coactivator
    • Li X, Monks B, Ge Q, Birnbaum MJ. Akt/PKB regulates hepatic metabolism by directly inhibiting PGC-1alpha transcription coactivator. Nature 2007; 447: 1012-1016.
    • (2007) Nature , vol.447 , pp. 1012-1016
    • Li, X.1    Monks, B.2    Ge, Q.3    Birnbaum, M.J.4
  • 96
    • 34547545892 scopus 로고    scopus 로고
    • AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1alpha
    • Jager S, Handschin C, St-Pierre J, Spiegelman BM. AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1alpha. Proc Natl Acad Sci USA 2007; 104: 12017-12022.
    • (2007) Proc Natl Acad Sci USA , vol.104 , pp. 12017-12022
    • Jager, S.1    Handschin, C.2    St-Pierre, J.3    Spiegelman, B.M.4
  • 97
    • 14544282413 scopus 로고    scopus 로고
    • Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1
    • Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Puigserver P. Nutrient control of glucose homeostasis through a complex of PGC-1alpha and SIRT1. Nature 2005; 434: 113-118.
    • (2005) Nature , vol.434 , pp. 113-118
    • Rodgers, J.T.1    Lerin, C.2    Haas, W.3    Gygi, S.P.4    Spiegelman, B.M.5    Puigserver, P.6
  • 98
    • 34247259630 scopus 로고    scopus 로고
    • Metabolic control of muscle mitochondrial function and fatty acid oxidation through SIRT1/PGC-1alpha
    • Gerhart-Hines Z, Rodgers JT, Bare O, et al. Metabolic control of muscle mitochondrial function and fatty acid oxidation through SIRT1/PGC-1alpha. Embo J 2007; 26: 1913-1923.
    • (2007) Embo J , vol.26 , pp. 1913-1923
    • Gerhart-Hines, Z.1    Rodgers, J.T.2    Bare, O.3
  • 99
    • 70449122132 scopus 로고    scopus 로고
    • Foxo1 integrates insulin signaling with mitochondrial function in the liver
    • Cheng Z, Guo S, Copps K, et al. Foxo1 integrates insulin signaling with mitochondrial function in the liver. Nat Med 2009; 15: 1307-1311.
    • (2009) Nat Med , vol.15 , pp. 1307-1311
    • Cheng, Z.1    Guo, S.2    Copps, K.3
  • 100
    • 0037337666 scopus 로고    scopus 로고
    • Gene disruption discloses role of selenoprotein P in selenium delivery to target tissues
    • Schomburg L, Schweizer U, Holtmann B, Flohe L, Sendtner M, Kohrle J. Gene disruption discloses role of selenoprotein P in selenium delivery to target tissues. Biochem J 2003; 370: 397-402.
    • (2003) Biochem J , vol.370 , pp. 397-402
    • Schomburg, L.1    Schweizer, U.2    Holtmann, B.3    Flohe, L.4    Sendtner, M.5    Kohrle, J.6
  • 101
    • 0037515627 scopus 로고    scopus 로고
    • Deletion of selenoprotein P alters distribution of selenium in the mouse
    • Hill KE, Zhou J, McMahan WJ, et al. Deletion of selenoprotein P alters distribution of selenium in the mouse. J Biol Chem 2003; 278: 13640-13646.
    • (2003) J Biol Chem , vol.278 , pp. 13640-13646
    • Hill, K.E.1    Zhou, J.2    McMahan, W.J.3
  • 102
    • 36248988618 scopus 로고    scopus 로고
    • Stimulation of selenoprotein P promoter activity in hepatoma cells by FoxO1a transcription factor
    • Walter PL, Steinbrenner H, Barthel A, Klotz LO. Stimulation of selenoprotein P promoter activity in hepatoma cells by FoxO1a transcription factor. Biochem Biophys Res Commun 2008; 365: 316-321.
    • (2008) Biochem Biophys Res Commun , vol.365 , pp. 316-321
    • Walter, P.L.1    Steinbrenner, H.2    Barthel, A.3    Klotz, L.O.4
  • 103
    • 58149395124 scopus 로고    scopus 로고
    • Selenoprotein P expression is controlled through interaction of the coactivator PGC-1alpha with FoxO1a and hepatocyte nuclear factor 4alpha transcription factors
    • Speckmann B, Walter PL, Alili L, et al. Selenoprotein P expression is controlled through interaction of the coactivator PGC-1alpha with FoxO1a and hepatocyte nuclear factor 4alpha transcription factors. Hepatology 2008; 48:1998-2006.
    • (2008) Hepatology , vol.48 , pp. 1998-2006
    • Speckmann, B.1    Walter, P.L.2    Alili, L.3
  • 105
    • 33646067098 scopus 로고    scopus 로고
    • Involvement of selenoprotein P in protection of human astrocytes from oxidative damage
    • Steinbrenner H, Alili L, Bilgic E, Sies H, Brenneisen P. Involvement of selenoprotein P in protection of human astrocytes from oxidative damage. Free Radic Biol Med 2006; 40: 1513-1523.
    • (2006) Free Radic Biol Med , vol.40 , pp. 1513-1523
    • Steinbrenner, H.1    Alili, L.2    Bilgic, E.3    Sies, H.4    Brenneisen, P.5
  • 106
    • 34547135428 scopus 로고    scopus 로고
    • Role of FoxO1 in FFA-induced oxidative stress in adipocytes
    • Subauste AR, Burant CF. Role of FoxO1 in FFA-induced oxidative stress in adipocytes. Am J Physiol Endocrinol Metab 2007; 293: E159-E164.
    • (2007) Am J Physiol Endocrinol Metab , vol.293 , pp. 159-164
    • Subauste, A.R.1    Burant, C.F.2
  • 107
    • 33749331558 scopus 로고    scopus 로고
    • Differential expression of FOXO1 and FOXO3a confers resistance to oxidative cell death upon endometrial decidualization
    • Kajihara T, Jones M, Fusi L, et al. Differential expression of FOXO1 and FOXO3a confers resistance to oxidative cell death upon endometrial decidualization. Mol Endocrinol 2006; 20:2444-2455.
    • (2006) Mol Endocrinol , vol.20 , pp. 2444-2455
    • Kajihara, T.1    Jones, M.2    Fusi, L.3
  • 108
    • 19444365211 scopus 로고    scopus 로고
    • PGC-1alpha regulates the mitochondrial antioxidant defense system in vascular endothelial cells
    • Valle I, Alvarez-Barrientos A, Arza E, Lamas S, Monsalve M. PGC-1alpha regulates the mitochondrial antioxidant defense system in vascular endothelial cells. Cardiovasc Res 2005; 66: 562-573.
    • (2005) Cardiovasc Res , vol.66 , pp. 562-573
    • Valle, I.1    Alvarez-Barrientos, A.2    Arza, E.3    Lamas, S.4    Monsalve, M.5
  • 109
    • 33644749330 scopus 로고    scopus 로고
    • Activation of AMP-activated protein kinase reduces hyperglycemia-induced mitochondrial reactive oxygen species production and promotes mitochondrial biogenesis in human umbilical vein endothelial cells
    • Kukidome D, Nishikawa T, Sonoda K, et al. Activation of AMP-activated protein kinase reduces hyperglycemia-induced mitochondrial reactive oxygen species production and promotes mitochondrial biogenesis in human umbilical vein endothelial cells. Diabetes 2006; 55: 120-127.
    • (2006) Diabetes , vol.55 , pp. 120-127
    • Kukidome, D.1    Nishikawa, T.2    Sonoda, K.3
  • 110
    • 33749999530 scopus 로고    scopus 로고
    • Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators
    • St-Pierre J, Drori S, Uldry M, et al. Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators. Cell 2006; 127: 397-408.
    • (2006) Cell , vol.127 , pp. 397-408
    • St-Pierre, J.1    Drori, S.2    Uldry, M.3
  • 111
    • 0038036024 scopus 로고    scopus 로고
    • Bioenergetic analysis of peroxisome proliferator-activated receptor gamma coactivators 1alpha and 1beta (PGC-1alpha and PGC-1beta) in muscle cells
    • St-Pierre J, Lin J, Krauss S, et al. Bioenergetic analysis of peroxisome proliferator-activated receptor gamma coactivators 1alpha and 1beta (PGC-1alpha and PGC-1beta) in muscle cells. J Biol Chem 2003; 278: 26597-26603.
    • (2003) J Biol Chem , vol.278 , pp. 26597-26603
    • St-Pierre, J.1    Lin, J.2    Krauss, S.3
  • 112
    • 67649819237 scopus 로고    scopus 로고
    • Mutual dependence of Foxo3a and PGC-1alpha in the induction of oxidative stress genes
    • Olmos Y, Valle I, Borniquel S, et al. Mutual dependence of Foxo3a and PGC-1alpha in the induction of oxidative stress genes. J Biol Chem 2009; 284: 14476-14484.
    • (2009) J Biol Chem , vol.284 , pp. 14476-14484
    • Olmos, Y.1    Valle, I.2    Borniquel, S.3
  • 113
    • 58349118928 scopus 로고    scopus 로고
    • Interactions between ROS and AMP kinase activity in the regulation of PGC-1alpha transcription in skeletal muscle cells
    • Irrcher I, Ljubicic V, Hood DA. Interactions between ROS and AMP kinase activity in the regulation of PGC-1alpha transcription in skeletal muscle cells. Am J Physiol Cell Physiol 2009; 296: C116-C123.
    • (2009) Am J Physiol Cell Physiol , vol.296 , pp. 116-123
    • Irrcher, I.1    Ljubicic, V.2    Hood, D.A.3
  • 114
    • 23944476164 scopus 로고    scopus 로고
    • Nutritional regulation of hepatic heme biosynthesis and porphyria through PGC-1alpha
    • Handschin C, Lin J, Rhee J, et al. Nutritional regulation of hepatic heme biosynthesis and porphyria through PGC-1alpha. Cell 2005; 122: 505-515.
    • (2005) Cell , vol.122 , pp. 505-515
    • Handschin, C.1    Lin, J.2    Rhee, J.3
  • 115
    • 0037404976 scopus 로고    scopus 로고
    • Recent advances in mechanisms regulating glucose oxidation at the level of the pyruvate dehydrogenase complex by PDKs
    • Sugden MC, Holness MJ. Recent advances in mechanisms regulating glucose oxidation at the level of the pyruvate dehydrogenase complex by PDKs. Am J Physiol Endocrinol Metab 2003; 284: E855-E862.
    • (2003) Am J Physiol Endocrinol Metab , vol.284 , pp. 855-862
    • Sugden, M.C.1    Holness, M.J.2
  • 116
    • 0034993356 scopus 로고    scopus 로고
    • Control of pyruvate dehydrogenase kinase gene expression
    • Harris RA, Huang B, Wu P. Control of pyruvate dehydrogenase kinase gene expression. Adv Enzyme Regul 2001; 41: 269-288.
    • (2001) Adv Enzyme Regul , vol.41 , pp. 269-288
    • Harris, R.A.1    Huang, B.2    Wu, P.3
  • 117
    • 0346158376 scopus 로고    scopus 로고
    • Regulation of pyruvate dehydrogenase complex activity by reversible phosphorylation
    • Holness MJ, Sugden MC. Regulation of pyruvate dehydrogenase complex activity by reversible phosphorylation. Biochem Soc Trans 2003; 31: 1143-1151.
    • (2003) Biochem Soc Trans , vol.31 , pp. 1143-1151
    • Holness, M.J.1    Sugden, M.C.2
  • 118
    • 1842475551 scopus 로고    scopus 로고
    • Protein kinase B-alpha inhibits human pyruvate dehydrogenase kinase-4 gene induction by dexamethasone through inactivation of FOXO transcription factors
    • Kwon HS, Huang B, Unterman TG, Harris RA. Protein kinase B-alpha inhibits human pyruvate dehydrogenase kinase-4 gene induction by dexamethasone through inactivation of FOXO transcription factors. Diabetes 2004; 53: 899-910.
    • (2004) Diabetes , vol.53 , pp. 899-910
    • Kwon, H.S.1    Huang, B.2    Unterman, T.G.3    Harris, R.A.4
  • 119
    • 23844470550 scopus 로고    scopus 로고
    • Cloning of the rat pyruvate dehydrogenase kinase 4 gene promoter: Activation of pyruvate dehydrogenase kinase 4 by the peroxisome proliferator-activated receptor gamma coactivator
    • Ma K, Zhang Y, Elam MB, Cook GA, Park EA. Cloning of the rat pyruvate dehydrogenase kinase 4 gene promoter: activation of pyruvate dehydrogenase kinase 4 by the peroxisome proliferator-activated receptor gamma coactivator. J Biol Chem 2005; 280: 29525-29532.
    • (2005) J Biol Chem , vol.280 , pp. 29525-29532
    • Ma, K.1    Zhang, Y.2    Elam, M.B.3    Cook, G.A.4    Park, E.A.5
  • 120
    • 0142200472 scopus 로고    scopus 로고
    • Forkhead transcription factor FOXO1 (FKHR)-dependent induction of PDK4 gene expression in skeletal muscle during energy deprivation
    • Furuyama T, Kitayama K, Yamashita H, Mori N. Forkhead transcription factor FOXO1 (FKHR)-dependent induction of PDK4 gene expression in skeletal muscle during energy deprivation. Biochem J 2003; 375: 365-371.
    • (2003) Biochem J , vol.375 , pp. 365-371
    • Furuyama, T.1    Kitayama, K.2    Yamashita, H.3    Mori, N.4
  • 121
    • 34247554887 scopus 로고    scopus 로고
    • Genome-wide orchestration of cardiac functions by the orphan nuclear receptors ERRalpha and gamma
    • Dufour CR, Wilson BJ, Huss JM, et al. Genome-wide orchestration of cardiac functions by the orphan nuclear receptors ERRalpha and gamma. Cell Metab 2007; 5: 345-356.
    • (2007) Cell Metab , vol.5 , pp. 345-356
    • Dufour, C.R.1    Wilson, B.J.2    Huss, J.M.3
  • 122
    • 33845973409 scopus 로고    scopus 로고
    • Estrogen-related receptors stimulate pyruvate dehydrogenase kinase isoform 4 gene expression
    • Zhang Y, Ma K, Sadana P, et al. Estrogen-related receptors stimulate pyruvate dehydrogenase kinase isoform 4 gene expression. J Biol Chem 2006; 281: 39897-39906.
    • (2006) J Biol Chem , vol.281 , pp. 39897-39906
    • Zhang, Y.1    Ma, K.2    Sadana, P.3
  • 123
    • 28544438180 scopus 로고    scopus 로고
    • PGC-1alpha coactivates PDK4 gene expression via the orphan nuclear receptor ERRalpha: A mechanism for transcriptional control of muscle glucose metabolism
    • Wende AR, Huss JM, Schaeffer PJ, Giguere V, Kelly DP. PGC-1alpha coactivates PDK4 gene expression via the orphan nuclear receptor ERRalpha: a mechanism for transcriptional control of muscle glucose metabolism. Mol Cell Biol 2005; 25: 10684-10694.
    • (2005) Mol Cell Biol , vol.25 , pp. 10684-10694
    • Wende, A.R.1    Huss, J.M.2    Schaeffer, P.J.3    Giguere, V.4    Kelly, D.P.5
  • 124
    • 71849109938 scopus 로고    scopus 로고
    • Regulation of pyruvate dehydrogenase kinase isoform 4 (PDK4) gene expression by glucocorticoids and insulin
    • Connaughton S, Chowdhury F, Attia RR, et al. Regulation of pyruvate dehydrogenase kinase isoform 4 (PDK4) gene expression by glucocorticoids and insulin. Mol Cell Endocrinol 2010; 315: 159-167.
    • (2010) Mol Cell Endocrinol , vol.315 , pp. 159-167
    • Connaughton, S.1    Chowdhury, F.2    Attia, R.R.3
  • 125
    • 2042425906 scopus 로고    scopus 로고
    • The IGF-1/PI3K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin ligases by inhibiting FOXO transcription factors
    • Stitt TN, Drujan D, Clarke BA, et al. The IGF-1/PI3K/Akt pathway prevents expression of muscle atrophy-induced ubiquitin ligases by inhibiting FOXO transcription factors. Mol Cell 2004;14: 395-403.
    • (2004) Mol Cell , vol.14 , pp. 395-403
    • Stitt, T.N.1    Drujan, D.2    Clarke, B.A.3
  • 126
    • 4544358547 scopus 로고    scopus 로고
    • Skeletal muscle FOXO1 (FKHR) transgenic mice have less skeletal muscle mass, down-regulated Type I (slow twitch/red muscle) fiber genes, and impaired glycemic control
    • Kamei Y, Miura S, Suzuki M, et al. Skeletal muscle FOXO1 (FKHR) transgenic mice have less skeletal muscle mass, down-regulated Type I (slow twitch/red muscle) fiber genes, and impaired glycemic control. J Biol Chem 2004; 279: 41114-41123.
    • (2004) J Biol Chem , vol.279 , pp. 41114-41123
    • Kamei, Y.1    Miura, S.2    Suzuki, M.3
  • 127
    • 33750697655 scopus 로고    scopus 로고
    • Epstein-Barr virus represses the FoxO1 transcription factor through latent membrane protein 1 and latent membrane protein 2A
    • Shore AM, White PC, Hui RC, et al. Epstein-Barr virus represses the FoxO1 transcription factor through latent membrane protein 1 and latent membrane protein 2A. J Virol 2006; 80: 11191-11199.
    • (2006) J Virol , vol.80 , pp. 11191-11199
    • Shore, A.M.1    White, P.C.2    Hui, R.C.3
  • 128
    • 41849150779 scopus 로고    scopus 로고
    • FOXOs, cancer and regulation of apoptosis
    • Fu Z, Tindall DJ. FOXOs, cancer and regulation of apoptosis. Oncogene 2008; 27: 2312-2319.
    • (2008) Oncogene , vol.27 , pp. 2312-2319
    • Fu, Z.1    Tindall, D.J.2
  • 131
    • 42449133390 scopus 로고    scopus 로고
    • Mechanisms of glucocorticoid-induced myopathy
    • Schakman O, Gilson H, Thissen JP. Mechanisms of glucocorticoid-induced myopathy. J Endocrinol 2008; 197: 1-10.
    • (2008) J Endocrinol , vol.197 , pp. 1-10
    • Schakman, O.1    Gilson, H.2    Thissen, J.P.3
  • 132
    • 56049123409 scopus 로고    scopus 로고
    • The glucocorticoid receptor and FOXO1 synergistically activate the skeletal muscle atrophy-associated MuRF1 gene
    • Waddell DS, Baehr LM, van den Brandt J, et al. The glucocorticoid receptor and FOXO1 synergistically activate the skeletal muscle atrophy-associated MuRF1 gene. Am J Physiol Endocrinol Metab 2008; 295: E785-E797.
    • (2008) Am J Physiol Endocrinol Metab , vol.295 , pp. 785-797
    • Waddell, D.S.1    Baehr, L.M.2    van den Brandt, J.3
  • 134
    • 0036087321 scopus 로고    scopus 로고
    • C/EBP DNA-binding activity is upregulated by a glucocorticoid-dependent mechanism in septic muscle
    • Penner G, Gang G, Sun X, Wray C, Hasselgren PO. C/EBP DNA-binding activity is upregulated by a glucocorticoid-dependent mechanism in septic muscle. Am J Physiol Regul Integr Comp Physiol 2002; 282: R439-R444.
    • (2002) Am J Physiol Regul Integr Comp Physiol , vol.282 , pp. 439-444
    • Penner, G.1    Gang, G.2    Sun, X.3    Wray, C.4    Hasselgren, P.O.5
  • 135
    • 20144365306 scopus 로고    scopus 로고
    • Expression and activity of C/EBPbeta and delta are upregulated by dexamethasone in skeletal muscle
    • Yang H, Mammen J, Wei W, et al. Expression and activity of C/EBPbeta and delta are upregulated by dexamethasone in skeletal muscle. J Cell Physiol 2005; 204: 219-226.
    • (2005) J Cell Physiol , vol.204 , pp. 219-226
    • Yang, H.1    Mammen, J.2    Wei, W.3
  • 136
    • 23944524470 scopus 로고    scopus 로고
    • Dexamethasone upregulates the expression of the nuclear cofactor p300 and its interaction with C/EBPbeta in cultured myotubes
    • Yang H, Menconi MJ, Wei W, Petkova V, Hasselgren PO. Dexamethasone upregulates the expression of the nuclear cofactor p300 and its interaction with C/EBPbeta in cultured myotubes. J Cell Biochem 2005; 94: 1058-1067.
    • (2005) J Cell Biochem , vol.94 , pp. 1058-1067
    • Yang, H.1    Menconi, M.J.2    Wei, W.3    Petkova, V.4    Hasselgren, P.O.5
  • 137
    • 57049104503 scopus 로고    scopus 로고
    • Foxo1 increases pro-inflammatory gene expression by inducing C/EBPbeta in TNF-alpha-treated adipocytes
    • Ito Y, Daitoku H, Fukamizu A. Foxo1 increases pro-inflammatory gene expression by inducing C/EBPbeta in TNF-alpha-treated adipocytes. Biochem Biophys Res Commun 2009; 378: 290-295.
    • (2009) Biochem Biophys Res Commun , vol.378 , pp. 290-295
    • Ito, Y.1    Daitoku, H.2    Fukamizu, A.3
  • 138
    • 33750825245 scopus 로고    scopus 로고
    • PGC-1alpha protects skeletal muscle from atrophy by suppressing FoxO3 action and atrophy-specific gene transcription
    • Sandri M, Lin J, Handschin C, et al. PGC-1alpha protects skeletal muscle from atrophy by suppressing FoxO3 action and atrophy-specific gene transcription. Proc Natl Acad Sci U S A. 2006; 103: 16260-16265.
    • (2006) Proc Natl Acad Sci U S A , vol.103 , pp. 16260-16265
    • Sandri, M.1    Lin, J.2    Handschin, C.3
  • 139
    • 9644270401 scopus 로고    scopus 로고
    • Atrogin-1/muscle atrophy F-box inhibits calcineurin-dependent cardiac hypertrophy by participating in an SCF ubiquitin ligase complex
    • Li HH, Kedar V, Zhang C, et al. Atrogin-1/muscle atrophy F-box inhibits calcineurin-dependent cardiac hypertrophy by participating in an SCF ubiquitin ligase complex. J Clin Invest. 2004; 114: 1058-1071.
    • (2004) J Clin Invest , vol.114 , pp. 1058-1071
    • Li, H.H.1    Kedar, V.2    Zhang, C.3
  • 140
    • 36049026136 scopus 로고    scopus 로고
    • Atrogin-1 inhibits Akt-dependent cardiac hypertrophy in mice via ubiquitin-dependent coactivation of Forkhead proteins
    • Li HH, Willis MS, Lockyer P, et al. Atrogin-1 inhibits Akt-dependent cardiac hypertrophy in mice via ubiquitin-dependent coactivation of Forkhead proteins. J Clin Invest 2007; 117: 3211-3223.
    • (2007) J Clin Invest , vol.117 , pp. 3211-3223
    • Li, H.H.1    Willis, M.S.2    Lockyer, P.3
  • 141
    • 33748675304 scopus 로고    scopus 로고
    • Foxo transcription factors blunt cardiac hypertrophy by inhibiting calcineurin signaling
    • Ni YG, Berenji K, Wang N, et al. Foxo transcription factors blunt cardiac hypertrophy by inhibiting calcineurin signaling. Circulation 2006; 114: 1159-1168.
    • (2006) Circulation , vol.114 , pp. 1159-1168
    • Ni, Y.G.1    Berenji, K.2    Wang, N.3
  • 142
    • 3943054839 scopus 로고    scopus 로고
    • The Sir2 family of protein deacetylases
    • Blander G, Guarente L. The Sir2 family of protein deacetylases. Annu Rev Biochem 2004; 73: 417-435.
    • (2004) Annu Rev Biochem , vol.73 , pp. 417-435
    • Blander, G.1    Guarente, L.2
  • 143
    • 0034677535 scopus 로고    scopus 로고
    • Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase
    • Imai S, Armstrong CM, Kaeberlein M, Guarente L. Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature 2000; 403: 795-800.
    • (2000) Nature , vol.403 , pp. 795-800
    • Imai, S.1    Armstrong, C.M.2    Kaeberlein, M.3    Guarente, L.4
  • 144
    • 0035826271 scopus 로고    scopus 로고
    • Increased dosage of a sir-2 gene extends lifespan in Caenorhabditis elegans
    • Tissenbaum HA, Guarente L. Increased dosage of a sir-2 gene extends lifespan in Caenorhabditis elegans. Nature 2001; 410: 227-230.
    • (2001) Nature , vol.410 , pp. 227-230
    • Tissenbaum, H.A.1    Guarente, L.2
  • 145
    • 0033600176 scopus 로고    scopus 로고
    • Characterization of five human cDNAs with homology to the yeast SIR2 gene: Sir2-like proteins (sirtuins) metabolize NAD and may have protein ADP-ribosyltransferase activity
    • Frye RA. Characterization of five human cDNAs with homology to the yeast SIR2 gene: Sir2-like proteins (sirtuins) metabolize NAD and may have protein ADP-ribosyltransferase activity. Biochem Biophys Res Commun 1999; 260: 273-279.
    • (1999) Biochem Biophys Res Commun , vol.260 , pp. 273-279
    • Frye, R.A.1
  • 146
    • 20144365700 scopus 로고    scopus 로고
    • Nuclear trapping of the forkhead transcription factor FoxO1 via Sirt-dependent deacetylationpromotes expression of glucogenetic genes
    • Frescas D, Valenti L, Accili D. Nuclear trapping of the forkhead transcription factor FoxO1 via Sirt-dependent deacetylationpromotes expression of glucogenetic genes. J Biol Chem 2005 280: 20589-20595.
    • (2005) J Biol Chem , vol.280 , pp. 20589-20595
    • Frescas, D.1    Valenti, L.2    Accili, D.3
  • 147
    • 71449105535 scopus 로고    scopus 로고
    • FoxO1 and HNF-4 are involved in regulation of hepatic glucokinase gene expression by resveratrol
    • Ganjam GK, Dimova EY, Unterman TG, Kietzmann T. FoxO1 and HNF-4 are involved in regulation of hepatic glucokinase gene expression by resveratrol. J Biol Chem 2009; 284: 30783-30797.
    • (2009) J Biol Chem , vol.284 , pp. 30783-30797
    • Ganjam, G.K.1    Dimova, E.Y.2    Unterman, T.G.3    Kietzmann, T.4
  • 148
    • 1342264308 scopus 로고    scopus 로고
    • Mammalian SIRT1 represses forkhead transcription factors
    • Motta MC, Divecha N, Lemieux M, et al. Mammalian SIRT1 represses forkhead transcription factors. Cell 2004; 116: 551-563.
    • (2004) Cell , vol.116 , pp. 551-563
    • Motta, M.C.1    Divecha, N.2    Lemieux, M.3
  • 150
    • 34249669270 scopus 로고    scopus 로고
    • Sirt1 regulates aging and resistance to oxidative stress in the heart
    • Alcendor RR, Gao S, Zhai P, et al. Sirt1 regulates aging and resistance to oxidative stress in the heart. Circ Res 2007; 100: 1512-1521.
    • (2007) Circ Res , vol.100 , pp. 1512-1521
    • Alcendor, R.R.1    Gao, S.2    Zhai, P.3
  • 151
    • 35348980724 scopus 로고    scopus 로고
    • SIRT1 controls endothelial angiogenic functions during vascular growth
    • Potente M, Ghaeni L, Baldessari D, et al. SIRT1 controls endothelial angiogenic functions during vascular growth. Genes Dev 2007; 21: 2644-2658.
    • (2007) Genes Dev , vol.21 , pp. 2644-2658
    • Potente, M.1    Ghaeni, L.2    Baldessari, D.3
  • 152
    • 33846005165 scopus 로고    scopus 로고
    • A novel class of vascular endothelial growth factor-responsive genes that require forkhead activity for expression
    • Abid MR, Shih SC, Otu HH, et al. A novel class of vascular endothelial growth factor-responsive genes that require forkhead activity for expression. J Biol Chem 2006; 281: 35544-35553.
    • (2006) J Biol Chem , vol.281 , pp. 35544-35553
    • Abid, M.R.1    Shih, S.C.2    Otu, H.H.3
  • 153
    • 34447626095 scopus 로고    scopus 로고
    • SIRT2 deacetylates FOXO3a in response to oxidative stress and caloric restriction
    • Wang F, Nguyen M, Qin FX, Tong Q. SIRT2 deacetylates FOXO3a in response to oxidative stress and caloric restriction. Aging Cell 2007; 6: 505-514.
    • (2007) Aging Cell , vol.6 , pp. 505-514
    • Wang, F.1    Nguyen, M.2    Qin, F.X.3    Tong, Q.4
  • 154
    • 10744232772 scopus 로고    scopus 로고
    • Variability of the SIRT3 gene, human silent information regulator Sir2 homologue, and survivorship in the elderly
    • Rose G, Dato S, Altomare K, et al. Variability of the SIRT3 gene, human silent information regulator Sir2 homologue, and survivorship in the elderly. Exp Gerontol 2003; 38: 1065-1070.
    • (2003) Exp Gerontol , vol.38 , pp. 1065-1070
    • Rose, G.1    Dato, S.2    Altomare, K.3
  • 155
    • 19944433088 scopus 로고    scopus 로고
    • A novel VNTR enhancer within the SIRT3 gene, a human homologue of SIR2, is associated with survival at oldest ages
    • Bellizzi D, Rose G, Cavalcante P, et al. A novel VNTR enhancer within the SIRT3 gene, a human homologue of SIR2, is associated with survival at oldest ages. Genomics 2005; 85: 258-263.
    • (2005) Genomics , vol.85 , pp. 258-263
    • Bellizzi, D.1    Rose, G.2    Cavalcante, P.3
  • 156
    • 0037135972 scopus 로고    scopus 로고
    • The human silent information regulator (Sir)2 homologue hSIRT3 is a mitochondrial nicotinamide adenine dinucleotide-dependent deacetylase
    • Schwer B, North BJ, Frye RA, Ott M, Verdin E. The human silent information regulator (Sir)2 homologue hSIRT3 is a mitochondrial nicotinamide adenine dinucleotide-dependent deacetylase. J Cell Biol. 2002; 158: 647-657.
    • (2002) J Cell Biol , vol.158 , pp. 647-657
    • Schwer, B.1    North, B.J.2    Frye, R.A.3    Ott, M.4    Verdin, E.5
  • 157
    • 34247271282 scopus 로고    scopus 로고
    • SirT3 is a nuclear NAD+-dependent histone deacetylase that translocates to the mitochondria upon cellular stress
    • Scher MB, Vaquero A, Reinberg D. SirT3 is a nuclear NAD+-dependent histone deacetylase that translocates to the mitochondria upon cellular stress. Genes Dev 2007; 21: 920-928.
    • (2007) Genes Dev , vol.21 , pp. 920-928
    • Scher, M.B.1    Vaquero, A.2    Reinberg, D.3
  • 158
    • 51449083112 scopus 로고    scopus 로고
    • SIRT3 interacts with the daf-16 homolog FOXO3a in the mitochondria, as well as increases FOXO3a dependent gene expression
    • Jacobs KM, Pennington JD, Bisht KS, et al. SIRT3 interacts with the daf-16 homolog FOXO3a in the mitochondria, as well as increases FOXO3a dependent gene expression. Int J Biol Sci 2008; 4:2 91-99.
    • (2008) Int J Biol Sci , vol.4 , Issue.2 , pp. 91-99
    • Jacobs, K.M.1    Pennington, J.D.2    Bisht, K.S.3
  • 159
    • 70349208608 scopus 로고    scopus 로고
    • Sirt3 blocks the cardiac hypertrophic response by augmenting Foxo3a-dependent antioxidant defense mechanisms in mice
    • Sundaresan NR, Gupta M, Kim G, Rajamohan SB, Isbatan A, Gupta MP. Sirt3 blocks the cardiac hypertrophic response by augmenting Foxo3a-dependent antioxidant defense mechanisms in mice. J Clin Invest 2009; 119: 2758-2771.
    • (2009) J Clin Invest , vol.119 , pp. 2758-2771
    • Sundaresan, N.R.1    Gupta, M.2    Kim, G.3    Rajamohan, S.B.4    Isbatan, A.5    Gupta, M.P.6
  • 160
    • 0037376913 scopus 로고    scopus 로고
    • Glucagon and regulation of glucose metabolism
    • Jiang G, Zhang BB. Glucagon and regulation of glucose metabolism. Am J Physiol Endocrinol Metab 2003; 284: E671-E678.
    • (2003) Am J Physiol Endocrinol Metab , vol.284 , pp. 671-678
    • Jiang, G.1    Zhang, B.B.2
  • 161
    • 27144506185 scopus 로고    scopus 로고
    • The CREB coactivator TORC2 is a key regulator of fasting glucose metabolism
    • Koo SH, Flechner L, Qi L, et al. The CREB coactivator TORC2 is a key regulator of fasting glucose metabolism. Nature 2005; 437: 1109-1111.
    • (2005) Nature , vol.437 , pp. 1109-1111
    • Koo, S.H.1    Flechner, L.2    Qi, L.3
  • 162
    • 0035855905 scopus 로고    scopus 로고
    • CREB regulates hepatic gluconeogenesis through the coactivator PGC-1
    • Herzig S, Long F, Jhala US, et al. CREB regulates hepatic gluconeogenesis through the coactivator PGC-1. Nature. 2001; 413: 179-183.
    • (2001) Nature , vol.413 , pp. 179-183
    • Herzig, S.1    Long, F.2    Jhala, U.S.3
  • 163
    • 56249100986 scopus 로고    scopus 로고
    • A fasting inducible switch modulates gluconeogenesis via activator/coactivator exchange
    • Liu Y, Dentin R, Chen D, et al. A fasting inducible switch modulates gluconeogenesis via activator/coactivator exchange. Nature 2008; 456: 269-273.
    • (2008) Nature , vol.456 , pp. 269-273
    • Liu, Y.1    Dentin, R.2    Chen, D.3
  • 164
    • 5444264003 scopus 로고    scopus 로고
    • Biological control through regulated transcriptional coactivators
    • Spiegelman BM, Heinrich R. Biological control through regulated transcriptional coactivators. Cell 2004; 119: 157-167.
    • (2004) Cell , vol.119 , pp. 157-167
    • Spiegelman, B.M.1    Heinrich, R.2
  • 165
    • 12944335299 scopus 로고    scopus 로고
    • DAF-16 recruits the CREB-binding protein coactivator complex to the insulin-like growth factor binding protein 1 promoter in HepG2 cells
    • Nasrin N, Ogg S, Cahill CM, et al. DAF-16 recruits the CREB-binding protein coactivator complex to the insulin-like growth factor binding protein 1 promoter in HepG2 cells. Proc Natl Acad Sci USA 2000; 97: 10412-10417.
    • (2000) Proc Natl Acad Sci USA , vol.97 , pp. 10412-10417
    • Nasrin, N.1    Ogg, S.2    Cahill, C.M.3
  • 166
    • 24344499875 scopus 로고    scopus 로고
    • The coactivator p300 directly acetylates the forkhead transcription factor Foxo1 and stimulates Foxo1-induced transcription
    • Perrot V, Rechler MM. The coactivator p300 directly acetylates the forkhead transcription factor Foxo1 and stimulates Foxo1-induced transcription. Mol Endocrinol 2005; 19: 2283-2298.
    • (2005) Mol Endocrinol , vol.19 , pp. 2283-2298
    • Perrot, V.1    Rechler, M.M.2
  • 167
    • 85047695408 scopus 로고    scopus 로고
    • Phosphorylation of forkhead transcription factors by erythropoietin and stem cell factor prevents acetylation and their interaction with coactivator p300 in erythroid progenitor cells
    • Mahmud DL, M GA, Deb DK, Platanias LC, Uddin S, Wickrema A. Phosphorylation of forkhead transcription factors by erythropoietin and stem cell factor prevents acetylation and their interaction with coactivator p300 in erythroid progenitor cells. Oncogene 2002; 21: 1556-1562.
    • (2002) Oncogene , vol.21 , pp. 1556-1562
    • Mahmud, D.L.1    Ga, M.2    Deb, D.K.3    Platanias, L.C.4    Uddin, S.5    Wickrema, A.6
  • 168
    • 2342445654 scopus 로고    scopus 로고
    • Negative regulation of forkhead transcription factor AFX (Foxo4) by CBP-induced acetylation
    • Fukuoka M, Daitoku H, Hatta M, Matsuzaki H, Umemura S, Fukamizu A. Negative regulation of forkhead transcription factor AFX (Foxo4) by CBP-induced acetylation. Int J Mol Med 2003; 12: 503-508.
    • (2003) Int J Mol Med , vol.12 , pp. 503-508
    • Fukuoka, M.1    Daitoku, H.2    Hatta, M.3    Matsuzaki, H.4    Umemura, S.5    Fukamizu, A.6
  • 169
    • 24744469490 scopus 로고    scopus 로고
    • SIRT1 is critical regulator of FOXO-mediated transcription in response to oxidative stress
    • Kobayashi Y, Furukawa-Hibi Y, Chen C, et al. SIRT1 is critical regulator of FOXO-mediated transcription in response to oxidative stress. Int J Mol Med 2005; 16: 237-243.
    • (2005) Int J Mol Med , vol.16 , pp. 237-243
    • Kobayashi, Y.1    Furukawa-Hibi, Y.2    Chen, C.3
  • 170
    • 3042750643 scopus 로고    scopus 로고
    • Silent information regulator 2 potentiates Foxo1-mediated transcription through its deacetylase activity
    • Daitoku H, Hatta M, Matsuzaki H, et al. Silent information regulator 2 potentiates Foxo1-mediated transcription through its deacetylase activity. Proc Natl Acad Sci USA 2004; 101: 10042-10047.
    • (2004) Proc Natl Acad Sci USA , vol.101 , pp. 10042-10047
    • Daitoku, H.1    Hatta, M.2    Matsuzaki, H.3
  • 171
    • 34547397081 scopus 로고    scopus 로고
    • SIRT2 regulates adipocyte differentiation through FoxO1 acetylation/deacetylation
    • Jing E, Gesta S, Kahn CR. SIRT2 regulates adipocyte differentiation through FoxO1 acetylation/deacetylation. Cell Metab 2007; 6: 105-114.
    • (2007) Cell Metab , vol.6 , pp. 105-114
    • Jing, E.1    Gesta, S.2    Kahn, C.R.3
  • 173
    • 58949091250 scopus 로고    scopus 로고
    • Acetylation curtails nucleosome binding, not stable nucleosome remodeling, by FoxO1
    • Hatta M, Liu F, Cirillo LA. Acetylation curtails nucleosome binding, not stable nucleosome remodeling, by FoxO1. Biochem Biophys Res Commun 2009; 379:1005-1008.
    • (2009) Biochem Biophys Res Commun , vol.379 , pp. 1005-1008
    • Hatta, M.1    Liu, F.2    Cirillo, L.A.3
  • 174
    • 16344384026 scopus 로고    scopus 로고
    • Suppression of FOXO1 activity by FHL2 through SIRT1-mediated deacetylation
    • Yang Y, Hou H, Haller EM, Nicosia SV, Bai W. Suppression of FOXO1 activity by FHL2 through SIRT1-mediated deacetylation. EMBO J 2005; 24: 1021-1032.
    • (2005) EMBO J , vol.24 , pp. 1021-1032
    • Yang, Y.1    Hou, H.2    Haller, E.M.3    Nicosia, S.V.4    Bai, W.5
  • 175
    • 69249229450 scopus 로고    scopus 로고
    • Redox-sensitive cysteines bridge p300/CBP-mediated acetylation and FoxO4 activity
    • Dansen TB, Smits LM, van Triest MH, et al. Redox-sensitive cysteines bridge p300/CBP-mediated acetylation and FoxO4 activity. Nat Chem Biol 2009; 5: 664-72.
    • (2009) Nat Chem Biol , vol.5 , pp. 664-672
    • Dansen, T.B.1    Smits, L.M.2    van Triest, M.H.3
  • 176
    • 0019026208 scopus 로고
    • Glycogen synthase kinase-3 from rabbit skeletal muscle. Separation from cyclic-AMP-dependent protein kinase and phosphorylase kinase
    • Embi N, Rylatt DB, Cohen P. Glycogen synthase kinase-3 from rabbit skeletal muscle. Separation from cyclic-AMP-dependent protein kinase and phosphorylase kinase. Eur J Biochem 1980; 107: 519-527.
    • (1980) Eur J Biochem , vol.107 , pp. 519-527
    • Embi, N.1    Rylatt, D.B.2    Cohen, P.3
  • 177
    • 0026353474 scopus 로고
    • CDNA cloning and properties of glycogen synthase kinase-3
    • Woodgett JR. CDNA cloning and properties of glycogen synthase kinase-3. Methods Enzymol 1991; 200: 564-577.
    • (1991) Methods Enzymol , vol.200 , pp. 564-577
    • Woodgett, J.R.1
  • 178
    • 0025286104 scopus 로고
    • Molecular cloning and expression of glycogen synthase kinase-3/factor A
    • Woodgett JR. Molecular cloning and expression of glycogen synthase kinase-3/factor A. Embo J 1990; 9: 2431-2438.
    • (1990) Embo J , vol.9 , pp. 2431-2438
    • Woodgett, J.R.1
  • 179
    • 0029587224 scopus 로고
    • Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B
    • Cross DA, Alessi DR, Cohen P, Andjelkovich M, Hemmings BA. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature 1995; 378: 785-789.
    • (1995) Nature , vol.378 , pp. 785-789
    • Cross, D.A.1    Alessi, D.R.2    Cohen, P.3    Andjelkovich, M.4    Hemmings, B.A.5
  • 180
    • 0034461612 scopus 로고    scopus 로고
    • Cyclic AMP promotes neuronal survival by phosphorylation of glycogen synthase kinase 3beta
    • Li M, Wang X, Meintzer MK, Laessig T, Birnbaum MJ, Heidenreich KA. Cyclic AMP promotes neuronal survival by phosphorylation of glycogen synthase kinase 3beta. Mol Cell Biol 2000; 20: 9356-9363.
    • (2000) Mol Cell Biol , vol.20 , pp. 9356-9363
    • Li, M.1    Wang, X.2    Meintzer, M.K.3    Laessig, T.4    Birnbaum, M.J.5    Heidenreich, K.A.6
  • 182
    • 0027430039 scopus 로고
    • Glycogen synthase kinase-3 is rapidly inactivated in response to insulin and phosphorylates eukaryotic initiation factor eIF-2B
    • Welsh GI, Proud CG. Glycogen synthase kinase-3 is rapidly inactivated in response to insulin and phosphorylates eukaryotic initiation factor eIF-2B. Biochem J 1993; 294: 625-629.
    • (1993) Biochem J , vol.294 , pp. 625-629
    • Welsh, G.I.1    Proud, C.G.2
  • 183
    • 0032498112 scopus 로고    scopus 로고
    • Regulation of eukaryotic initiation factor eIF2B: Glycogen synthase kinase-3 phosphorylates a conserved serine which undergoes dephosphorylation in response to insulin
    • Welsh GI, Miller CM, Loughlin AJ, Price NT, Proud CG. Regulation of eukaryotic initiation factor eIF2B: glycogen synthase kinase-3 phosphorylates a conserved serine which undergoes dephosphorylation in response to insulin. FEBS Lett 1998; 421: 125-130.
    • (1998) FEBS Lett , vol.421 , pp. 125-130
    • Welsh, G.I.1    Miller, C.M.2    Loughlin, A.J.3    Price, N.T.4    Proud, C.G.5
  • 184
    • 0033971454 scopus 로고    scopus 로고
    • Biochemical interactions in the wnt pathway
    • Seidensticker MJ, Behrens J. Biochemical interactions in the wnt pathway. Biochim Biophys Acta 2000; 1495: 168-182.
    • (2000) Biochim Biophys Acta , vol.1495 , pp. 168-182
    • Seidensticker, M.J.1    Behrens, J.2
  • 185
    • 0033851276 scopus 로고    scopus 로고
    • GSK3, a master switch regulating cell-fate specification and tumorigenesis
    • Kim L, Kimmel AR. GSK3, a master switch regulating cell-fate specification and tumorigenesis. Curr Opin Genet Dev 2000; 10: 508-514.
    • (2000) Curr Opin Genet Dev , vol.10 , pp. 508-514
    • Kim, L.1    Kimmel, A.R.2
  • 187
    • 33845355511 scopus 로고    scopus 로고
    • Diversity of LEF/TCF action in development and disease
    • Arce L, Yokoyama NN, Waterman ML. Diversity of LEF/TCF action in development and disease. Oncogene 2006; 25: 7492-7504.
    • (2006) Oncogene , vol.25 , pp. 7492-7504
    • Arce, L.1    Yokoyama, N.N.2    Waterman, M.L.3
  • 188
    • 0035477020 scopus 로고    scopus 로고
    • GSK3 takes centre stage more than 20 years after its discovery
    • Frame S, Cohen P. GSK3 takes centre stage more than 20 years after its discovery. Biochem J 2001; 359: 1-16.
    • (2001) Biochem J , vol.359 , pp. 1-16
    • Frame, S.1    Cohen, P.2
  • 189
    • 66249108601 scopus 로고    scopus 로고
    • Understanding the Warburg effect: The metabolic requirements of cell proliferation
    • Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 2009; 324: 1029-1033.
    • (2009) Science , vol.324 , pp. 1029-1033
    • Vander, H.M.G.1    Cantley, L.C.2    Thompson, C.B.3
  • 191
    • 40349098058 scopus 로고    scopus 로고
    • Glycogen synthase kinase 3 (GSK3) in the heart: A point of integration in hypertrophic signalling and a therapeutic target? A critical analysis
    • Sugden PH, Fuller SJ, Weiss SC, Clerk A. Glycogen synthase kinase 3 (GSK3) in the heart: a point of integration in hypertrophic signalling and a therapeutic target? A critical analysis. Br J Pharmacol 2008; 153: S137-S153.
    • (2008) Br J Pharmacol , vol.153 , pp. 137-153
    • Sugden, P.H.1    Fuller, S.J.2    Weiss, S.C.3    Clerk, A.4
  • 192
    • 37849041966 scopus 로고    scopus 로고
    • Forkhead transcription factors and cardiovascular biology
    • Papanicolaou KN, Izumiya Y, Walsh K. Forkhead transcription factors and cardiovascular biology. Circ Res 2008; 102: 16-31.
    • (2008) Circ Res , vol.102 , pp. 16-31
    • Papanicolaou, K.N.1    Izumiya, Y.2    Walsh, K.3
  • 193
    • 33846295218 scopus 로고    scopus 로고
    • FoxOs are lineage-restricted redundant tumor suppressors and regulate endothelial cell homeostasis
    • Paik JH, Kollipara R, Chu G, et al. FoxOs are lineage-restricted redundant tumor suppressors and regulate endothelial cell homeostasis. Cell 2007; 128: 309-323.
    • (2007) Cell , vol.128 , pp. 309-323
    • Paik, J.H.1    Kollipara, R.2    Chu, G.3
  • 194
    • 34447504987 scopus 로고    scopus 로고
    • Perspectives on endothelial-to-mesenchymal transition: Potential contribution to vascular remodeling in chronic pulmonary hypertension
    • Arciniegas E, Frid MG, Douglas IS, Stenmark KR. Perspectives on endothelial-to-mesenchymal transition: potential contribution to vascular remodeling in chronic pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2007; 293: L1-8.
    • (2007) Am J Physiol Lung Cell Mol Physiol , vol.293 , pp. 1-8
    • Arciniegas, E.1    Frid, M.G.2    Douglas, I.S.3    Stenmark, K.R.4
  • 195
    • 3042588831 scopus 로고    scopus 로고
    • Molecular regulation of vascular smooth muscle cell differentiation in development and disease
    • Owens GK, Kumar MS, Wamhoff BR. Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol Rev 2004 J; 84: 767-801.
    • Physiol Rev 2004 J , vol.84 , pp. 767-801
    • Owens, G.K.1    Kumar, M.S.2    Wamhoff, B.R.3
  • 196
    • 34249736457 scopus 로고    scopus 로고
    • Programming smooth muscle plasticity with chromatin dynamics
    • McDonald OG, Owens GK. Programming smooth muscle plasticity with chromatin dynamics. Circ Res 2007; 100: 1428-1441.
    • (2007) Circ Res , vol.100 , pp. 1428-1441
    • McDonald, O.G.1    Owens, G.K.2
  • 197
    • 33745152386 scopus 로고    scopus 로고
    • The myocardin family of transcriptional coactivators: Versatile regulators of cell growth, migration, and myogenesis
    • Pipes GC, Creemers EE, Olson EN. The myocardin family of transcriptional coactivators: versatile regulators of cell growth, migration, and myogenesis. Genes Dev 2006; 20: 1545-1556.
    • (2006) Genes Dev , vol.20 , pp. 1545-1556
    • Pipes, G.C.1    Creemers, E.E.2    Olson, E.N.3
  • 198
    • 0027425113 scopus 로고
    • Activation of ternary complex factor Elk-1 by MAP kinases
    • Janknecht R, Ernst WH, Pingoud V, Nordheim A. Activation of ternary complex factor Elk-1 by MAP kinases. Embo J 1993; 12: 5097-5104.
    • (1993) Embo J , vol.12 , pp. 5097-5104
    • Janknecht, R.1    Ernst, W.H.2    Pingoud, V.3    Nordheim, A.4
  • 199
    • 1642297200 scopus 로고    scopus 로고
    • Myocardin and ternary complex factors compete for SRF to control smooth muscle gene expression
    • Wang Z, Wang DZ, Hockemeyer D, McAnally J, Nordheim A, Olson EN. Myocardin and ternary complex factors compete for SRF to control smooth muscle gene expression. Nature 2004; 428: 185-189.
    • (2004) Nature , vol.428 , pp. 185-189
    • Wang, Z.1    Wang, D.Z.2    Hockemeyer, D.3    McAnally, J.4    Nordheim, A.5    Olson, E.N.6
  • 200
    • 0038737042 scopus 로고    scopus 로고
    • Actin dynamics control SRF activity by regulation of its coactivator MAL
    • Miralles F, Posern G, Zaromytidou AI, Treisman R. Actin dynamics control SRF activity by regulation of its coactivator MAL. Cell 2003; 113: 329-342.
    • (2003) Cell , vol.113 , pp. 329-342
    • Miralles, F.1    Posern, G.2    Zaromytidou, A.I.3    Treisman, R.4
  • 201
    • 0032582808 scopus 로고    scopus 로고
    • Differentiated phenotype of smooth muscle cells depends on signaling pathways through insulin-like growth factors and phosphatidylinositol 3-kinase
    • Hayashi K, Saga H, Chimori Y, Kimura K, Yamanaka Y, Sobue K. Differentiated phenotype of smooth muscle cells depends on signaling pathways through insulin-like growth factors and phosphatidylinositol 3-kinase. J Biol Chem 1998; 273: 28860-28867.
    • (1998) J Biol Chem , vol.273 , pp. 28860-28867
    • Hayashi, K.1    Saga, H.2    Chimori, Y.3    Kimura, K.4    Yamanaka, Y.5    Sobue, K.6
  • 202
    • 22944451474 scopus 로고    scopus 로고
    • Phenotypic modulation of smooth muscle cells through interaction of Foxo4 and myocardin
    • Liu ZP, Wang Z, Yanagisawa H, Olson EN. Phenotypic modulation of smooth muscle cells through interaction of Foxo4 and myocardin. Dev Cell 2005; 9: 261-270.
    • (2005) Dev Cell , vol.9 , pp. 261-270
    • Liu, Z.P.1    Wang, Z.2    Yanagisawa, H.3    Olson, E.N.4
  • 203
    • 23844491841 scopus 로고    scopus 로고
    • Forkhead transcription factors inhibit vascular smooth muscle cell proliferation and neointimal hyperplasia
    • Abid MR, Yano K, Guo S, et al. Forkhead transcription factors inhibit vascular smooth muscle cell proliferation and neointimal hyperplasia. J Biol Chem 2005; 280: 29864-29873.
    • (2005) J Biol Chem , vol.280 , pp. 29864-29873
    • Abid, M.R.1    Yano, K.2    Guo, S.3
  • 204
    • 0042413919 scopus 로고    scopus 로고
    • Mechanosensitive p27Kip1 regulation and cell cycle entry in vascular smooth muscle cells
    • Sedding DG, Seay U, Fink L, et al. Mechanosensitive p27Kip1 regulation and cell cycle entry in vascular smooth muscle cells. Circulation 2003; 108: 616-622.
    • (2003) Circulation , vol.108 , pp. 616-622
    • Sedding, D.G.1    Seay, U.2    Fink, L.3
  • 205
    • 0141483026 scopus 로고    scopus 로고
    • Myocardin mRNA is augmented in the failing myocardium: Expression profiling in the porcine model and human dilated cardiomyopathy
    • Torrado M, Lopez E, Centeno A, Medrano C, Castro-Beiras A, Mikhailov AT. Myocardin mRNA is augmented in the failing myocardium: expression profiling in the porcine model and human dilated cardiomyopathy. J Mol Med 2003; 81: 566-577.
    • (2003) J Mol Med , vol.81 , pp. 566-577
    • Torrado, M.1    Lopez, E.2    Centeno, A.3    Medrano, C.4    Castro-Beiras, A.5    Mikhailov, A.T.6
  • 206
    • 33646798121 scopus 로고    scopus 로고
    • Myocardin induces cardiomyocyte hypertrophy
    • Xing W, Zhang TC, Cao D, et al. Myocardin induces cardiomyocyte hypertrophy. Circ Res 2006; 98: 1089-1097.
    • (2006) Circ Res , vol.98 , pp. 1089-1097
    • Xing, W.1    Zhang, T.C.2    Cao, D.3
  • 207
    • 26244440041 scopus 로고    scopus 로고
    • Glycogen synthase kinase 3beta inhibits myocardin-dependent transcription and hypertrophy induction through site-specific phosphorylation
    • Badorff C, Seeger FH, Zeiher AM, Dimmeler S. Glycogen synthase kinase 3beta inhibits myocardin-dependent transcription and hypertrophy induction through site-specific phosphorylation. Circ Res 2005; 97: 645-654.
    • (2005) Circ Res , vol.97 , pp. 645-654
    • Badorff, C.1    Seeger, F.H.2    Zeiher, A.M.3    Dimmeler, S.4
  • 208
    • 33751534932 scopus 로고    scopus 로고
    • Myocardin is a direct transcriptional target of Mef2, Tead and Foxo proteins during cardiovascular development
    • Creemers EE, Sutherland LB, McAnally J, Richardson JA, Olson EN. Myocardin is a direct transcriptional target of Mef2, Tead and Foxo proteins during cardiovascular development. Development 2006; 133: 4245-4256.
    • (2006) Development , vol.133 , pp. 4245-4256
    • Creemers, E.E.1    Sutherland, L.B.2    McAnally, J.3    Richardson, J.A.4    Olson, E.N.5
  • 209
    • 34147195312 scopus 로고    scopus 로고
    • FoxO4 regulates tumor necrosis factor alpha-directed smooth muscle cell migration by activating matrix metalloproteinase 9 gene transcription
    • Li H, Liang J, Castrillon DH, DePinho RA, Olson EN, Liu ZP. FoxO4 regulates tumor necrosis factor alpha-directed smooth muscle cell migration by activating matrix metalloproteinase 9 gene transcription. Mol Cell Biol 2007; 27: 2676-2686.
    • (2007) Mol Cell Biol , vol.27 , pp. 2676-2686
    • Li, H.1    Liang, J.2    Castrillon, D.H.3    Depinho, R.A.4    Olson, E.N.5    Liu, Z.P.6
  • 210
    • 63649158684 scopus 로고    scopus 로고
    • FoxO1 inhibits leptin regulation of pro-opiomelanocortin promoter activity by blocking STAT3 interaction with specificity protein 1
    • Yang G, Lim CY, Li C, et al. FoxO1 inhibits leptin regulation of pro-opiomelanocortin promoter activity by blocking STAT3 interaction with specificity protein 1. J Biol Chem 2009; 284: 3719-3727.
    • (2009) J Biol Chem , vol.284 , pp. 3719-3727
    • Yang, G.1    Lim, C.Y.2    Li, C.3
  • 211
    • 36849053911 scopus 로고    scopus 로고
    • Dual roles of myocardin-related transcription factors in epithelial mesenchymal transition via slug induction and actin remodeling
    • Morita T, Mayanagi T, Sobue K. Dual roles of myocardin-related transcription factors in epithelial mesenchymal transition via slug induction and actin remodeling. J Cell Biol 2007; 179: 1027-1042.
    • (2007) J Cell Biol , vol.179 , pp. 1027-1042
    • Morita, T.1    Mayanagi, T.2    Sobue, K.3
  • 212
    • 69249208738 scopus 로고    scopus 로고
    • Regulation of myocardin-related transcriptional coactivators through cofactor interactions in differentiation and cancer
    • Brandt DT, Xu J, Steinbeisser H, Grosse R. Regulation of myocardin-related transcriptional coactivators through cofactor interactions in differentiation and cancer. Cell Cycle 2009; 8: 2523-2527.
    • (2009) Cell Cycle , vol.8 , pp. 2523-2527
    • Brandt, D.T.1    Xu, J.2    Steinbeisser, H.3    Grosse, R.4
  • 213
    • 51049114512 scopus 로고    scopus 로고
    • Rho/Rho-associated kinase signal regulates myogenic differentiation via myocardin-related transcription factor-A/Smad-dependent transcription of the Id3 gene
    • Iwasaki K, Hayashi K, Fujioka T, Sobue K. Rho/Rho-associated kinase signal regulates myogenic differentiation via myocardin-related transcription factor-A/Smad-dependent transcription of the Id3 gene. J Biol Chem 2008; 283: 21230-21241.
    • (2008) J Biol Chem , vol.283 , pp. 21230-21241
    • Iwasaki, K.1    Hayashi, K.2    Fujioka, T.3    Sobue, K.4
  • 214
    • 27944454815 scopus 로고    scopus 로고
    • Bone morphogenetic protein signaling modulates myocardin transactivation of cardiac genes
    • Callis TE, Cao D, Wang DZ. Bone morphogenetic protein signaling modulates myocardin transactivation of cardiac genes. Circ Res 2005; 97: 992-1000.
    • (2005) Circ Res , vol.97 , pp. 992-1000
    • Callis, T.E.1    Cao, D.2    Wang, D.Z.3
  • 215
    • 27944445459 scopus 로고    scopus 로고
    • Myocardin enhances Smad3-mediated transforming growth factor-beta1 signaling in a CArG box-independent manner: Smad-binding element is an important cis element for SM22alpha transcription in vivo
    • Qiu P, Ritchie RP, Fu Z, et al. Myocardin enhances Smad3-mediated transforming growth factor-beta1 signaling in a CArG box-independent manner: Smad-binding element is an important cis element for SM22alpha transcription in vivo. Circ Res 2005; 97: 983-991.
    • (2005) Circ Res , vol.97 , pp. 983-991
    • Qiu, P.1    Ritchie, R.P.2    Fu, Z.3
  • 216
    • 33644857012 scopus 로고    scopus 로고
    • Increased Wnt signaling triggers oncogenic conversion of human breast epithelial cells by a Notch-dependent mechanism
    • Ayyanan A, Civenni G, Ciarloni L, et al. Increased Wnt signaling triggers oncogenic conversion of human breast epithelial cells by a Notch-dependent mechanism. Proc Natl Acad Sci USA 2006; 103: 3799-3804.
    • (2006) Proc Natl Acad Sci USA , vol.103 , pp. 3799-3804
    • Ayyanan, A.1    Civenni, G.2    Ciarloni, L.3
  • 217
    • 27144504699 scopus 로고    scopus 로고
    • The Notch pathway in ovarian carcinomas and adenomas
    • Hopfer O, Zwahlen D, Fey MF, Aebi S. The Notch pathway in ovarian carcinomas and adenomas. Br J Cancer 2005; 93:709-718.
    • (2005) Br J Cancer , vol.93 , pp. 709-718
    • Hopfer, O.1    Zwahlen, D.2    Fey, M.F.3    Aebi, S.4
  • 218
    • 7244250027 scopus 로고    scopus 로고
    • Loss of negative regulation by Numb over Notch is relevant to human breast carcinogenesis
    • Pece S, Serresi M, Santolini E, et al. Loss of negative regulation by Numb over Notch is relevant to human breast carcinogenesis. J Cell Biol 2004; 167: 215-221.
    • (2004) J Cell Biol , vol.167 , pp. 215-221
    • Pece, S.1    Serresi, M.2    Santolini, E.3
  • 219
    • 0036734148 scopus 로고    scopus 로고
    • Activation of Notch-1 signaling maintains the neoplastic phenotype in human Ras-transformed cells
    • Weijzen S, Rizzo P, Braid M, et al. Activation of Notch-1 signaling maintains the neoplastic phenotype in human Ras-transformed cells. Nat Med 2002; 8: 979-986.
    • (2002) Nat Med , vol.8 , pp. 979-986
    • Weijzen, S.1    Rizzo, P.2    Braid, M.3
  • 220
    • 0025856717 scopus 로고
    • TAN-1, the human homolog of the Drosophila notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms
    • Ellisen LW, Bird J, West DC, et al. TAN-1, the human homolog of the Drosophila notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms. Cell 1991; 66: 649-661.
    • (1991) Cell , vol.66 , pp. 649-661
    • Ellisen, L.W.1    Bird, J.2    West, D.C.3
  • 221
    • 0942287279 scopus 로고    scopus 로고
    • Embryonic myogenesis pathways in muscle regeneration
    • Zhao P, Hoffman EP. Embryonic myogenesis pathways in muscle regeneration. Dev Dyn 2004; 229: 380-392.
    • (2004) Dev Dyn , vol.229 , pp. 380-392
    • Zhao, P.1    Hoffman, E.P.2
  • 222
    • 24344443668 scopus 로고    scopus 로고
    • The regulation of Notch signaling in muscle stem cell activation and postnatal myogenesis
    • Luo D, Renault VM, Rando TA. The regulation of Notch signaling in muscle stem cell activation and postnatal myogenesis. Semin Cell Dev Biol 2005; 16: 612-622.
    • (2005) Semin Cell Dev Biol , vol.16 , pp. 612-622
    • Luo, D.1    Renault, V.M.2    Rando, T.A.3
  • 223
    • 0032915458 scopus 로고    scopus 로고
    • Notch signaling imposes two distinct blocks in the differentiation of C2C12 myoblasts
    • Nofziger D, Miyamoto A, Lyons KM, Weinmaster G. Notch signaling imposes two distinct blocks in the differentiation of C2C12 myoblasts. Development 1999; 126: 1689-1702.
    • (1999) Development , vol.126 , pp. 1689-1702
    • Nofziger, D.1    Miyamoto, A.2    Lyons, K.M.3    Weinmaster, G.4
  • 224
    • 0033548489 scopus 로고    scopus 로고
    • Delta-induced Notch signaling mediated by RBP-J inhibits MyoD expression and myogenesis
    • Kuroda K, Tani S, Tamura K, Minoguchi S, Kurooka H, Honjo T. Delta-induced Notch signaling mediated by RBP-J inhibits MyoD expression and myogenesis. J Biol Chem 1999; 274: 7238-7244.
    • (1999) J Biol Chem , vol.274 , pp. 7238-7244
    • Kuroda, K.1    Tani, S.2    Tamura, K.3    Minoguchi, S.4    Kurooka, H.5    Honjo, T.6
  • 225
    • 11144356337 scopus 로고    scopus 로고
    • Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy
    • Sandri M, Sandri C, Gilbert A, et al. Foxo transcription factors induce the atrophy-related ubiquitin ligase atrogin-1 and cause skeletal muscle atrophy. Cell 2004; 117: 399-412.
    • (2004) Cell , vol.117 , pp. 399-412
    • Sandri, M.1    Sandri, C.2    Gilbert, A.3
  • 226
    • 0042769210 scopus 로고    scopus 로고
    • Forkhead transcription factor FoxO1 transduces insulin-like growth factor's signal to p27Kip1 in primary skeletal muscle satellite cells
    • Machida S, Spangenburg EE, Booth FW. Forkhead transcription factor FoxO1 transduces insulin-like growth factor's signal to p27Kip1 in primary skeletal muscle satellite cells. J Cell Physiol 2003; 196: 523-531.
    • (2003) J Cell Physiol , vol.196 , pp. 523-531
    • Machida, S.1    Spangenburg, E.E.2    Booth, F.W.3
  • 227
    • 0037416153 scopus 로고    scopus 로고
    • FKHR (FOXO1a) is required for myotube fusion of primary mouse myoblasts
    • Bois PR, Grosveld GC. FKHR (FOXO1a) is required for myotube fusion of primary mouse myoblasts. Embo J 2003; 22: 1147-1157.
    • (2003) Embo J , vol.22 , pp. 1147-1157
    • Bois, P.R.1    Grosveld, G.C.2
  • 228
    • 34848838902 scopus 로고    scopus 로고
    • A Foxo/Notch pathway controls myogenic differentiation and fiber type specification
    • Kitamura T, Kitamura YI, Funahashi Y, et al. A Foxo/Notch pathway controls myogenic differentiation and fiber type specification. J Clin Invest 2007; 117: 2477-2485.
    • (2007) J Clin Invest , vol.117 , pp. 2477-2485
    • Kitamura, T.1    Kitamura, Y.I.2    Funahashi, Y.3
  • 229
    • 34447125200 scopus 로고    scopus 로고
    • P53 homologue, p51/p63, maintains the immaturity of keratinocyte stem cells by inhibiting Notch1 activity
    • Okuyama R, Ogawa E, Nagoshi H, et al. P53 homologue, p51/p63, maintains the immaturity of keratinocyte stem cells by inhibiting Notch1 activity. Oncogene 2007; 26: 4478-4488.
    • (2007) Oncogene , vol.26 , pp. 4478-4488
    • Okuyama, R.1    Ogawa, E.2    Nagoshi, H.3
  • 230
    • 22344436360 scopus 로고    scopus 로고
    • P21WAF1/Cip1 is a negative transcriptional regulator of Wnt4 expression downstream of Notch1 activation
    • Devgan V, Mammucari C, Millar SE, Brisken C, Dotto GP. P21WAF1/Cip1 is a negative transcriptional regulator of Wnt4 expression downstream of Notch1 activation. Genes Dev 2005; 19: 1485-1495.
    • (2005) Genes Dev , vol.19 , pp. 1485-1495
    • Devgan, V.1    Mammucari, C.2    Millar, S.E.3    Brisken, C.4    Dotto, G.P.5
  • 231
    • 0037370310 scopus 로고    scopus 로고
    • Notch1 functions as a tumor suppressor in mouse skin
    • Nicolas M, Wolfer A, Raj K, et al. Notch1 functions as a tumor suppressor in mouse skin. Nat Genet 2003; 33: 416-421.
    • (2003) Nat Genet , vol.33 , pp. 416-421
    • Nicolas, M.1    Wolfer, A.2    Raj, K.3
  • 232
    • 17844384528 scopus 로고    scopus 로고
    • Notch signaling is a direct determinant of keratinocyte growth arrest and entry into differentiation
    • Rangarajan A, Talora C, Okuyama R, et al. Notch signaling is a direct determinant of keratinocyte growth arrest and entry into differentiation. Embo J 2001; 20: 3427-3436.
    • (2001) Embo J , vol.20 , pp. 3427-3436
    • Rangarajan, A.1    Talora, C.2    Okuyama, R.3
  • 233
    • 33947166199 scopus 로고    scopus 로고
    • Notch1 is a p53 target gene involved in human keratinocyte tumor suppression through negative regulation of ROCK1/2 and MRCKalpha kinases
    • Lefort K, Mandinova A, Ostano P, et al. Notch1 is a p53 target gene involved in human keratinocyte tumor suppression through negative regulation of ROCK1/2 and MRCKalpha kinases. Genes Dev 2007; 21: 562-577.
    • (2007) Genes Dev , vol.21 , pp. 562-577
    • Lefort, K.1    Mandinova, A.2    Ostano, P.3
  • 234
    • 42449147521 scopus 로고    scopus 로고
    • The FoxO3a gene is a key negative target of canonical Notch signalling in the keratinocyte UVB response
    • Mandinova A, Lefort K, Tommasi di Vignano A, et al. The FoxO3a gene is a key negative target of canonical Notch signalling in the keratinocyte UVB response. Embo J 2008; 27: 1243-1254.
    • (2008) Embo J , vol.27 , pp. 1243-1254
    • Mandinova, A.1    Lefort, K.2    Di Vignano, T.A.3
  • 237
    • 33845808721 scopus 로고    scopus 로고
    • Myogenic progenitor cells in the mouse embryo are marked by the expression of Pax3/7 genes that regulate their survival and myogenic potential
    • Buckingham M, Bajard L, Daubas P, et al. Myogenic progenitor cells in the mouse embryo are marked by the expression of Pax3/7 genes that regulate their survival and myogenic potential. Anat Embryol (Berl) 2006; 211: 51-56.
    • (2006) Anat Embryol (Berl) , vol.211 , pp. 51-56
    • Buckingham, M.1    Bajard, L.2    Daubas, P.3
  • 238
    • 0030891318 scopus 로고    scopus 로고
    • Ectopic Pax-3 activates MyoD and Myf-5 expression in embryonic mesoderm and neural tissue
    • Maroto M, Reshef R, Munsterberg AE, Koester S, Goulding M, Lassar AB. Ectopic Pax-3 activates MyoD and Myf-5 expression in embryonic mesoderm and neural tissue. Cell 1997; 89: 139-148.
    • (1997) Cell , vol.89 , pp. 139-148
    • Maroto, M.1    Reshef, R.2    Munsterberg, A.E.3    Koester, S.4    Goulding, M.5    Lassar, A.B.6
  • 239
    • 0028205002 scopus 로고
    • Pax-3 is required for the development of limb muscles: A possible role for the migration of dermomyotomal muscle progenitor cells
    • Bober E, Franz T, Arnold HH, Gruss P, Tremblay P. Pax-3 is required for the development of limb muscles: a possible role for the migration of dermomyotomal muscle progenitor cells. Development 1994; 120: 603-612.
    • (1994) Development , vol.120 , pp. 603-612
    • Bober, E.1    Franz, T.2    Arnold, H.H.3    Gruss, P.4    Tremblay, P.5
  • 240
    • 33847309514 scopus 로고    scopus 로고
    • Fusions involving PAX and FOX genes in the molecular pathogenesis of alveolar rhabdomyosarcoma: Recent advances
    • Mercado GE, Barr FG. Fusions involving PAX and FOX genes in the molecular pathogenesis of alveolar rhabdomyosarcoma: recent advances. Curr Mol Med 2007; 7: 47-61.
    • (2007) Curr Mol Med , vol.7 , pp. 47-61
    • Mercado, G.E.1    Barr, F.G.2
  • 241
    • 53249095869 scopus 로고    scopus 로고
    • Codependent activators direct myoblast-specific MyoD transcription
    • Hu P, Geles KG, Paik JH, DePinho RA, Tjian R. Codependent activators direct myoblast-specific MyoD transcription. Dev Cell 2008; 15: 534-546.
    • (2008) Dev Cell , vol.15 , pp. 534-546
    • Hu, P.1    Geles, K.G.2    Paik, J.H.3    Depinho, R.A.4    Tjian, R.5
  • 242
    • 0242499448 scopus 로고    scopus 로고
    • Cytostatic and apoptotic actions of TGF-beta in homeostasis and cancer
    • Siegel PM, Massague J. Cytostatic and apoptotic actions of TGF-beta in homeostasis and cancer. Nat Rev Cancer 2003; 3: 807-821.
    • (2003) Nat Rev Cancer , vol.3 , pp. 807-821
    • Siegel, P.M.1    Massague, J.2
  • 243
    • 23044466047 scopus 로고    scopus 로고
    • Specificity and versatility in tgf-beta signaling through Smads
    • Feng XH, Derynck R. Specificity and versatility in tgf-beta signaling through Smads. Annu Rev Cell Dev Biol 2005; 21: 659-693.
    • (2005) Annu Rev Cell Dev Biol , vol.21 , pp. 659-693
    • Feng, X.H.1    Derynck, R.2
  • 245
    • 47849127561 scopus 로고    scopus 로고
    • Fibrosis in diabetes complications: Pathogenic mechanisms and circulating and urinary markers
    • Ban CR, Twigg SM. Fibrosis in diabetes complications: pathogenic mechanisms and circulating and urinary markers. Vasc Health Risk Manag 2008; 4: 575-596.
    • (2008) Vasc Health Risk Manag , vol.4 , pp. 575-596
    • Ban, C.R.1    Twigg, S.M.2
  • 246
    • 34748828509 scopus 로고    scopus 로고
    • Obesity and obesity-initiated metabolic syndrome: Mechanistic links to chronic kidney disease
    • Wahba IM, Mak RH. Obesity and obesity-initiated metabolic syndrome: mechanistic links to chronic kidney disease. Clin J Am Soc Nephrol 2007; 2: 550-562.
    • (2007) Clin J Am Soc Nephrol , vol.2 , pp. 550-562
    • Wahba, I.M.1    Mak, R.H.2
  • 247
    • 57349083077 scopus 로고    scopus 로고
    • Transcriptional regulation of the plasminogen activator inhibitor type 1--with an emphasis on negative regulation
    • Nagamine Y. Transcriptional regulation of the plasminogen activator inhibitor type 1--with an emphasis on negative regulation. Thromb Haemost 2008; 100: 1007-1013.
    • (2008) Thromb Haemost , vol.100 , pp. 1007-1013
    • Nagamine, Y.1
  • 248
    • 20244376914 scopus 로고    scopus 로고
    • Transforming growth factor-beta and platelet-derived growth factor signal via c-Jun N-terminal kinase-dependent Smad2/3 phosphorylation in rat hepatic stellate cells after acute liver injury
    • Yoshida K, Matsuzaki K, Mori S, Tahashi Y, Yamagata H, Furukawa F, et al. Transforming growth factor-beta and platelet-derived growth factor signal via c-Jun N-terminal kinase-dependent Smad2/3 phosphorylation in rat hepatic stellate cells after acute liver injury. Am J Pathol 2005; 166: 1029-1039.
    • (2005) Am J Pathol , vol.166 , pp. 1029-1039
    • Yoshida, K.1    Matsuzaki, K.2    Mori, S.3    Tahashi, Y.4    Yamagata, H.5    Furukawa, F.6
  • 249
    • 0028838983 scopus 로고
    • Induction of plasminogen activator inhibitor type-1 (PAI-1) by proinsulin and insulin in vivo
    • Nordt TK, Sawa H, Fujii S, Sobel BE. Induction of plasminogen activator inhibitor type-1 (PAI-1) by proinsulin and insulin in vivo. Circulation 1995; 91: 764-770.
    • (1995) Circulation , vol.91 , pp. 764-770
    • Nordt, T.K.1    Sawa, H.2    Fujii, S.3    Sobel, B.E.4
  • 250
    • 67650433374 scopus 로고    scopus 로고
    • Forkhead transcription factor FoxO1 inhibits insulin-and transforming growth factor-beta-stimulated plasminogen activator inhibitor-1 expression
    • Jung YA, Lee KM, Kim MK, Jung GS, Seo YJ, Kim HS, et al. Forkhead transcription factor FoxO1 inhibits insulin-and transforming growth factor-beta-stimulated plasminogen activator inhibitor-1 expression. Biochem Biophys Res Commun 2009; 386: 757-761.
    • (2009) Biochem Biophys Res Commun , vol.386 , pp. 757-761
    • Jung, Y.A.1    Lee, K.M.2    Kim, M.K.3    Jung, G.S.4    Seo, Y.J.5    Kim, H.S.6
  • 251
    • 0030776478 scopus 로고    scopus 로고
    • The DAF-3 Smad protein antagonizes TGF-beta-related receptor signaling in the Caenorhabditis elegans dauer pathway
    • Patterson GI, Koweek A, Wong A, Liu Y, Ruvkun G. The DAF-3 Smad protein antagonizes TGF-beta-related receptor signaling in the Caenorhabditis elegans dauer pathway. Genes Dev 1997; 11: 2679-2690.
    • (1997) Genes Dev , vol.11 , pp. 2679-2690
    • Patterson, G.I.1    Koweek, A.2    Wong, A.3    Liu, Y.4    Ruvkun, G.5
  • 252
    • 10544256181 scopus 로고    scopus 로고
    • Control of C. elegans larval development by neuronal expression of a TGF-beta homolog
    • Ren P, Lim CS, Johnsen R, Albert PS, Pilgrim D, Riddle DL. Control of C. elegans larval development by neuronal expression of a TGF-beta homolog. Science 1996; 274: 1389-1391.
    • (1996) Science , vol.274 , pp. 1389-1391
    • Ren, P.1    Lim, C.S.2    Johnsen, R.3    Albert, P.S.4    Pilgrim, D.5    Riddle, D.L.6
  • 253
    • 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
  • 254
    • 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
  • 255
    • 33646562542 scopus 로고    scopus 로고
    • The logic of TGFbeta signaling
    • Massague J, Gomis RR. The logic of TGFbeta signaling. FEBS Lett 2006; 580: 2811-2820.
    • (2006) FEBS Lett , vol.580 , pp. 2811-2820
    • Massague, J.1    Gomis, R.R.2
  • 256
    • 33748208668 scopus 로고    scopus 로고
    • C/EBPbeta at the core of the TGFbeta cytostatic response and its evasion in metastatic breast cancer cells
    • Gomis RR, Alarcon C, Nadal C, Van Poznak C, Massague J. C/EBPbeta at the core of the TGFbeta cytostatic response and its evasion in metastatic breast cancer cells. Cancer Cell 2006; 10: 203-214.
    • (2006) Cancer Cell , vol.10 , pp. 203-214
    • Gomis, R.R.1    Alarcon, C.2    Nadal, C.3    van Poznak, C.4    Massague, J.5
  • 257
    • 33748075143 scopus 로고    scopus 로고
    • A FoxO-Smad synexpression group in human keratinocytes
    • Gomis RR, Alarcon C, He W, et al. A FoxO-Smad synexpression group in human keratinocytes. Proc Natl Acad Sci USA 2006; 103: 12747-12752.
    • (2006) Proc Natl Acad Sci USA , vol.103 , pp. 12747-12752
    • Gomis, R.R.1    Alarcon, C.2    He, W.3
  • 258
    • 0037039312 scopus 로고    scopus 로고
    • CCAAT/enhancer binding protein-beta is a mediator of keratinocyte survival and skin tumorigenesis involving oncogenic Ras signaling
    • Zhu S, Yoon K, Sterneck E, Johnson PF, Smart RC. CCAAT/enhancer binding protein-beta is a mediator of keratinocyte survival and skin tumorigenesis involving oncogenic Ras signaling. Proc Natl Acad Sci USA 2002; 99: 207-212.
    • (2002) Proc Natl Acad Sci USA , vol.99 , pp. 207-212
    • Zhu, S.1    Yoon, K.2    Sterneck, E.3    Johnson, P.F.4    Smart, R.C.5
  • 259
    • 4344714027 scopus 로고    scopus 로고
    • Cell cycle-dependent phosphorylation of C/EBPbeta mediates oncogenic cooperativity between C/EBPbeta and H-RasV12
    • Shuman JD, Sebastian T, Kaldis P, et al. Cell cycle-dependent phosphorylation of C/EBPbeta mediates oncogenic cooperativity between C/EBPbeta and H-RasV12. Mol Cell Biol 2004; 24: 7380-7391.
    • (2004) Mol Cell Biol , vol.24 , pp. 7380-7391
    • Shuman, J.D.1    Sebastian, T.2    Kaldis, P.3
  • 260
    • 33846319709 scopus 로고    scopus 로고
    • Decreased survival of C/EBP beta-deficient keratinocytes is due to aberrant regulation of p53 levels and function
    • Yoon K, Zhu S, Ewing SJ, Smart RC. Decreased survival of C/EBP beta-deficient keratinocytes is due to aberrant regulation of p53 levels and function. Oncogene 2007; 26: 360-367.
    • (2007) Oncogene , vol.26 , pp. 360-367
    • Yoon, K.1    Zhu, S.2    Ewing, S.J.3    Smart, R.C.4
  • 261
    • 54049144611 scopus 로고    scopus 로고
    • C/EBPbeta represses p53 to promote cell survival downstream of DNA damage independent of oncogenic Ras and p19(Arf)
    • Ewing SJ, Zhu S, Zhu F, House JS, Smart RC. C/EBPbeta represses p53 to promote cell survival downstream of DNA damage independent of oncogenic Ras and p19(Arf). Cell Death Differ 2008; 15: 1734-1744.
    • (2008) Cell Death Differ , vol.15 , pp. 1734-1744
    • Ewing, S.J.1    Zhu, S.2    Zhu, F.3    House, J.S.4    Smart, R.C.5
  • 262
    • 0038322020 scopus 로고    scopus 로고
    • Transforming growth factor-beta inhibits adipocyte differentiation by Smad3 interacting with CCAAT/enhancer-binding protein (C/EBP) and repressing C/EBP transactivation function
    • Choy L, Derynck R. Transforming growth factor-beta inhibits adipocyte differentiation by Smad3 interacting with CCAAT/enhancer-binding protein (C/EBP) and repressing C/EBP transactivation function. J Biol Chem 2003; 278: 9609-9619.
    • (2003) J Biol Chem , vol.278 , pp. 9609-9619
    • Choy, L.1    Derynck, R.2
  • 263
    • 0035984289 scopus 로고    scopus 로고
    • Interaction between TGFbeta signaling proteins and C/EBP controls basal and Tat-mediated transcription of HIV-1 LTR in astrocytes
    • Coyle-Rink J, Sweet T, Abraham S, et al. Interaction between TGFbeta signaling proteins and C/EBP controls basal and Tat-mediated transcription of HIV-1 LTR in astrocytes. Virology 2002; 299: 240-247.
    • (2002) Virology , vol.299 , pp. 240-247
    • Coyle-Rink, J.1    Sweet, T.2    Abraham, S.3
  • 264
    • 0035816686 scopus 로고    scopus 로고
    • Smad proteins suppress CCAAT/enhancer-binding protein (C/EBP) beta-and STAT3-mediated transcriptional activation of the haptoglobin promoter
    • Zauberman A, Lapter S, Zipori D. Smad proteins suppress CCAAT/enhancer-binding protein (C/EBP) beta-and STAT3-mediated transcriptional activation of the haptoglobin promoter. J Biol Chem 2001; 276: 24719-24725.
    • (2001) J Biol Chem , vol.276 , pp. 24719-24725
    • Zauberman, A.1    Lapter, S.2    Zipori, D.3
  • 265
    • 67649202062 scopus 로고    scopus 로고
    • Forkhead box O-class 1 and forkhead box G1 as prognostic markers for bladder cancer
    • Kim TH, Jo SW, Lee YS, et al. Forkhead box O-class 1 and forkhead box G1 as prognostic markers for bladder cancer. J Korean Med Sci 2009; 24: 468-473.
    • (2009) J Korean Med Sci , vol.24 , pp. 468-473
    • Kim, T.H.1    Jo, S.W.2    Lee, Y.S.3
  • 267
    • 70349973752 scopus 로고    scopus 로고
    • Overexpression of FOXG1 contributes to TGF-beta resistance through inhibition of p21WAF1/CIP1 expression in ovarian cancer
    • Chan DW, Liu VW, To RM, et al. Overexpression of FOXG1 contributes to TGF-beta resistance through inhibition of p21WAF1/CIP1 expression in ovarian cancer. Br J Cancer 2009; 101: 1433-1443.
    • (2009) Br J Cancer , vol.101 , pp. 1433-1443
    • Chan, D.W.1    Liu, V.W.2    To, R.M.3
  • 268
    • 37349099564 scopus 로고    scopus 로고
    • Generation of Cajal-Retzius neurons in mouse forebrain is regulated by transforming growth factor beta-Fox signaling pathways
    • Siegenthaler JA, Miller MW. Generation of Cajal-Retzius neurons in mouse forebrain is regulated by transforming growth factor beta-Fox signaling pathways. Dev Biol 2008 313: 35-46
    • (2008) Dev Biol , vol.313 , pp. 35-46
    • Siegenthaler, J.A.1    Miller, M.W.2
  • 269
    • 37349016202 scopus 로고    scopus 로고
    • Kruppel-like factor 5 mediates cellular transformation during oncogenic KRAS-induced intestinal tumorigenesis
    • Nandan MO, McConnell BB, Ghaleb AM, et al. Kruppel-like factor 5 mediates cellular transformation during oncogenic KRAS-induced intestinal tumorigenesis. Gastroenterology. 2008; 134: 120-130.
    • (2008) Gastroenterology , vol.134 , pp. 120-130
    • Nandan, M.O.1    McConnell, B.B.2    Ghaleb, A.M.3
  • 270
    • 0037179848 scopus 로고    scopus 로고
    • A possible tumor suppressor role of the KLF5 transcription factor in human breast cancer
    • Chen C, Bhalala HV, Qiao H, Dong JT. A possible tumor suppressor role of the KLF5 transcription factor in human breast cancer. Oncogene 2002; 21: 6567-6572.
    • (2002) Oncogene , vol.21 , pp. 6567-6572
    • Chen, C.1    Bhalala, H.V.2    Qiao, H.3    Dong, J.T.4
  • 271
    • 1842425769 scopus 로고    scopus 로고
    • Intestinal tumor progression is associated with altered function of KLF5
    • Bateman NW, Tan D, Pestell RG, Black JD, Black AR. Intestinal tumor progression is associated with altered function of KLF5. J Biol Chem 2004; 279: 12093-12101.
    • (2004) J Biol Chem , vol.279 , pp. 12093-12101
    • Bateman, N.W.1    Tan, D.2    Pestell, R.G.3    Black, J.D.4    Black, A.R.5
  • 272
    • 65249114297 scopus 로고    scopus 로고
    • Pro-proliferative factor KLF5 becomes anti-proliferative in epithelial homeostasis upon signaling-mediated modification
    • Guo P, Dong XY, Zhang X, et al. Pro-proliferative factor KLF5 becomes anti-proliferative in epithelial homeostasis upon signaling-mediated modification. J Biol Chem 2009; 284: 6071-6078.
    • (2009) J Biol Chem , vol.284 , pp. 6071-6078
    • Guo, P.1    Dong, X.Y.2    Zhang, X.3
  • 273
    • 67650555438 scopus 로고    scopus 로고
    • Acetylation of KLF5 alters the assembly of p15 transcription factors in transforming growth factor-beta-mediated induction in epithelial cells
    • Guo P, Zhao KW, Dong XY, Sun X, Dong JT. Acetylation of KLF5 alters the assembly of p15 transcription factors in transforming growth factor-beta-mediated induction in epithelial cells. J Biol Chem 2009; 284: 18184-18193.
    • (2009) J Biol Chem , vol.284 , pp. 18184-18193
    • Guo, P.1    Zhao, K.W.2    Dong, X.Y.3    Sun, X.4    Dong, J.T.5
  • 274
    • 70350502092 scopus 로고    scopus 로고
    • Opposing effects of KLF5 on the transcription of MYC in epithelial proliferation in the context of transforming growth factor beta
    • Guo P, Dong XY, Zhao K, Sun X, Li Q, Dong JT. Opposing effects of KLF5 on the transcription of MYC in epithelial proliferation in the context of transforming growth factor beta. J Biol Chem 2009; 284: 28243-28252.
    • (2009) J Biol Chem , vol.284 , pp. 28243-28252
    • Guo, P.1    Dong, X.Y.2    Zhao, K.3    Sun, X.4    Li, Q.5    Dong, J.T.6
  • 275
  • 276
    • 53149108993 scopus 로고    scopus 로고
    • TGFbeta1, TNFalpha, and insulin signaling crosstalk in regulation of the rat cholesterol 7alpha-hydroxylase gene expression
    • Li T, Ma H, Chiang JY. TGFbeta1, TNFalpha, and insulin signaling crosstalk in regulation of the rat cholesterol 7alpha-hydroxylase gene expression. J Lipid Res 2008; 49: 1981-1989.
    • (2008) J Lipid Res , vol.49 , pp. 1981-1989
    • Li, T.1    Ma, H.2    Chiang, J.Y.3
  • 277
    • 0033869876 scopus 로고    scopus 로고
    • Translational control of C/EBPalpha and C/EBPbeta isoform expression
    • Calkhoven CF, Muller C, Leutz A. Translational control of C/EBPalpha and C/EBPbeta isoform expression. Genes Dev 2000; 14: 1920-1932.
    • (2000) Genes Dev , vol.14 , pp. 1920-1932
    • Calkhoven, C.F.1    Muller, C.2    Leutz, A.3
  • 278
    • 34547133494 scopus 로고    scopus 로고
    • The C/EBP family of transcription factors: A paradigm for interaction between gene expression and proliferation control
    • Nerlov C. The C/EBP family of transcription factors: a paradigm for interaction between gene expression and proliferation control. Trends Cell Biol 2007; 17: 318-324.
    • (2007) Trends Cell Biol , vol.17 , pp. 318-324
    • Nerlov, C.1
  • 279
    • 33751510219 scopus 로고    scopus 로고
    • Suppression of C/EBPalpha expression in periportal hepatoblasts may stimulate biliary cell differentiation through increased Hnf6 and Hnf1b expression
    • Yamasaki H, Sada A, Iwata T, et al. Suppression of C/EBPalpha expression in periportal hepatoblasts may stimulate biliary cell differentiation through increased Hnf6 and Hnf1b expression. Development 2006; 133: 4233-4243.
    • (2006) Development , vol.133 , pp. 4233-4243
    • Yamasaki, H.1    Sada, A.2    Iwata, T.3
  • 280
    • 0032504613 scopus 로고    scopus 로고
    • Hepatocytes deficient in CCAAT/enhancer binding protein alpha (C/EBP alpha) exhibit both hepatocyte and biliary epithelial cell character
    • Tomizawa M, Garfield S, Factor V, Xanthopoulos KG. Hepatocytes deficient in CCAAT/enhancer binding protein alpha (C/EBP alpha) exhibit both hepatocyte and biliary epithelial cell character. Biochem Biophys Res Commun 1998; 249: 1-5.
    • (1998) Biochem Biophys Res Commun , vol.249 , pp. 1-5
    • Tomizawa, M.1    Garfield, S.2    Factor, V.3    Xanthopoulos, K.G.4
  • 281
    • 0035896598 scopus 로고    scopus 로고
    • A nucleoprotein complex containing CCAAT/enhancer-binding protein beta interacts with an insulin response sequence in the insulin-like growth factor-binding protein-1 gene and contributes to insulin-regulated gene expression
    • Ghosh AK, Lacson R, Liu P, et al. A nucleoprotein complex containing CCAAT/enhancer-binding protein beta interacts with an insulin response sequence in the insulin-like growth factor-binding protein-1 gene and contributes to insulin-regulated gene expression. J Biol Chem 2001; 276: 8507-8515.
    • (2001) J Biol Chem , vol.276 , pp. 8507-8515
    • Ghosh, A.K.1    Lacson, R.2    Liu, P.3
  • 282
    • 34547824172 scopus 로고    scopus 로고
    • Foxo1 links insulin signaling to C/EBPalpha and regulates gluconeogenesis during liver development
    • Sekine K, Chen YR, Kojima N, Ogata K, Fukamizu A, Miyajima A. Foxo1 links insulin signaling to C/EBPalpha and regulates gluconeogenesis during liver development. Embo J 2007; 26: 3607-3615.
    • (2007) Embo J , vol.26 , pp. 3607-3615
    • Sekine, K.1    Chen, Y.R.2    Kojima, N.3    Ogata, K.4    Fukamizu, A.5    Miyajima, A.6
  • 283
    • 0028980389 scopus 로고
    • C/EBP alpha: A critical regulator of genes governing integrative metabolic processes
    • Darlington GJ, Wang N, Hanson RW. C/EBP alpha: a critical regulator of genes governing integrative metabolic processes. Curr Opin Genet Dev 1995; 5: 565-570.
    • (1995) Curr Opin Genet Dev , vol.5 , pp. 565-570
    • Darlington, G.J.1    Wang, N.2    Hanson, R.W.3
  • 284
    • 2642549921 scopus 로고    scopus 로고
    • Liver-enriched transcription factors in liver function and development. Part II: The C/EBPs and D site-binding protein in cell cycle control, carcinogenesis, circadian gene regulation, liver regeneration, apoptosis, and liver-specific gene regulation
    • Schrem H, Klempnauer J, Borlak J. Liver-enriched transcription factors in liver function and development. Part II: the C/EBPs and D site-binding protein in cell cycle control, carcinogenesis, circadian gene regulation, liver regeneration, apoptosis, and liver-specific gene regulation. Pharmacol Rev 2004; 56: 291-330.
    • (2004) Pharmacol Rev , vol.56 , pp. 291-330
    • Schrem, H.1    Klempnauer, J.2    Borlak, J.3
  • 285
    • 41549153181 scopus 로고    scopus 로고
    • C/EBPs: Recipients of extracellular signals through proteome modulation
    • Nerlov C. C/EBPs: recipients of extracellular signals through proteome modulation. Curr Opin Cell Biol 2008; 20: 180-185.
    • (2008) Curr Opin Cell Biol , vol.20 , pp. 180-185
    • Nerlov, C.1
  • 286
    • 51849147067 scopus 로고    scopus 로고
    • Role of FoxO1 activation in MDR1 expression in adriamycin-resistant breast cancer cells
    • Han CY, Cho KB, Choi HS, Han HK, Kang KW. Role of FoxO1 activation in MDR1 expression in adriamycin-resistant breast cancer cells. Carcinogenesis 2008; 29: 1837-1844.
    • (2008) Carcinogenesis , vol.29 , pp. 1837-1844
    • Han, C.Y.1    Cho, K.B.2    Choi, H.S.3    Han, H.K.4    Kang, K.W.5
  • 287
    • 0037036407 scopus 로고    scopus 로고
    • Cyclic AMP-induced forkhead transcription factor, FKHR, cooperates with CCAAT/enhancer-binding protein beta in differentiating human endometrial stromal cells
    • Christian M, Zhang X, Schneider-Merck T, et al. Cyclic AMP-induced forkhead transcription factor, FKHR, cooperates with CCAAT/enhancer-binding protein beta in differentiating human endometrial stromal cells. J Biol Chem 2002; 277: 20825-20832.
    • (2002) J Biol Chem , vol.277 , pp. 20825-20832
    • Christian, M.1    Zhang, X.2    Schneider-Merck, T.3
  • 288
    • 0030928627 scopus 로고    scopus 로고
    • Progesterone-dependent decidualization of the human endometrium is mediated by cAMP
    • Brar AK, Frank GR, Kessler CA, Cedars MI, Handwerger S. Progesterone-dependent decidualization of the human endometrium is mediated by cAMP. Endocrine 1997; 6: 301-307.
    • (1997) Endocrine , vol.6 , pp. 301-307
    • Brar, A.K.1    Frank, G.R.2    Kessler, C.A.3    Cedars, M.I.4    Handwerger, S.5
  • 289
    • 0033305291 scopus 로고    scopus 로고
    • Progesterone receptor regulates decidual prolactin expression in differentiating human endometrial stromal cells
    • Brosens JJ, Hayashi N, White JO. Progesterone receptor regulates decidual prolactin expression in differentiating human endometrial stromal cells. Endocrinology 1999; 140: 4809-4820.
    • (1999) Endocrinology , vol.140 , pp. 4809-4820
    • Brosens, J.J.1    Hayashi, N.2    White, J.O.3
  • 290
    • 0031047405 scopus 로고    scopus 로고
    • Activated protein kinase A is required for differentiation-dependent transcription of the decidual prolactin gene in human endometrial stromal cells
    • Telgmann R, Maronde E, Tasken K, Gellersen B. Activated protein kinase A is required for differentiation-dependent transcription of the decidual prolactin gene in human endometrial stromal cells. Endocrinology 1997; 138: 929-937.
    • (1997) Endocrinology , vol.138 , pp. 929-937
    • Telgmann, R.1    Maronde, E.2    Tasken, K.3    Gellersen, B.4
  • 291
    • 0033609858 scopus 로고    scopus 로고
    • CCAAT/enhancer-binding proteins are mediators in the protein kinase A-dependent activation of the decidual prolactin promoter
    • Pohnke Y, Kempf R, Gellersen B. CCAAT/enhancer-binding proteins are mediators in the protein kinase A-dependent activation of the decidual prolactin promoter. J Biol Chem 1999; 274: 24808-24818.
    • (1999) J Biol Chem , vol.274 , pp. 24808-24818
    • Pohnke, Y.1    Kempf, R.2    Gellersen, B.3
  • 292
    • 73849111344 scopus 로고    scopus 로고
    • Forkhead transcription factor Foxo1 is essential for adipocyte differentiation
    • Munekata K, Sakamoto K. Forkhead transcription factor Foxo1 is essential for adipocyte differentiation. In Vitro Cell Dev Biol Anim 2009; 45: 642-651.
    • (2009) In Vitro Cell Dev Biol Anim , vol.45 , pp. 642-651
    • Munekata, K.1    Sakamoto, K.2
  • 295
    • 0027515736 scopus 로고
    • Insulin-like growth factor (IGF), IGF binding protein (IGFBP), and IGF receptor gene expression and IGFBP synthesis in human uterine leiomyomata
    • Giudice LC, Irwin JC, Dsupin BA, et al. Insulin-like growth factor (IGF), IGF binding protein (IGFBP), and IGF receptor gene expression and IGFBP synthesis in human uterine leiomyomata. Hum Reprod 1993; 8: 1796-806.
    • (1993) Hum Reprod , vol.8 , pp. 1796-1806
    • Giudice, L.C.1    Irwin, J.C.2    Dsupin, B.A.3
  • 296
    • 0037235335 scopus 로고    scopus 로고
    • Regulation of insulin-like growth factor binding protein-1 promoter activity by FKHR and HOXA10 in primate endometrial cells
    • Kim JJ, Taylor HS, Akbas GE, et al. Regulation of insulin-like growth factor binding protein-1 promoter activity by FKHR and HOXA10 in primate endometrial cells. Biol Reprod 2003; 68: 24-30.
    • (2003) Biol Reprod , vol.68 , pp. 24-30
    • Kim, J.J.1    Taylor, H.S.2    Akbas, G.E.3
  • 297
    • 0029793280 scopus 로고    scopus 로고
    • Mechanisms of reduced fertility in Hoxa-10 mutant mice: Uterine homeosis and loss of maternal Hoxa-10 expression
    • Benson GV, Lim H, Paria BC, Satokata I, Dey SK, Maas RL. Mechanisms of reduced fertility in Hoxa-10 mutant mice: uterine homeosis and loss of maternal Hoxa-10 expression. Development 1996; 122: 2687-2696.
    • (1996) Development , vol.122 , pp. 2687-2696
    • Benson, G.V.1    Lim, H.2    Paria, B.C.3    Satokata, I.4    Dey, S.K.5    Maas, R.L.6
  • 298
    • 0030778154 scopus 로고    scopus 로고
    • Insulin-like growth factor binding protein-1: Recent findings and new directions
    • Lee PD, Giudice LC, Conover CA, Powell DR. Insulin-like growth factor binding protein-1: recent findings and new directions. Proc Soc Exp Biol Med 1997; 216: 319-357.
    • (1997) Proc Soc Exp Biol Med , vol.216 , pp. 319-357
    • Lee, P.D.1    Giudice, L.C.2    Conover, C.A.3    Powell, D.R.4
  • 299
    • 0034039896 scopus 로고    scopus 로고
    • Transgenic mouse models for studying the functions of insulin-like growth factor-binding proteins
    • Schneider MR, Lahm H, Wu M, Hoeflich A, Wolf E. Transgenic mouse models for studying the functions of insulin-like growth factor-binding proteins. Faseb J 2000; 14: 629-640.
    • (2000) Faseb J , vol.14 , pp. 629-640
    • Schneider, M.R.1    Lahm, H.2    Wu, M.3    Hoeflich, A.4    Wolf, E.5
  • 300
    • 0030999590 scopus 로고    scopus 로고
    • Liver-specific expression of human insulin-like growth factor binding protein-1 in transgenic mice: Repercussions on reproduction, ante-and perinatal mortality and postnatal growth
    • Gay E, Seurin D, Babajko S, Doublier S, Cazillis M, Binoux M. Liver-specific expression of human insulin-like growth factor binding protein-1 in transgenic mice: repercussions on reproduction, ante-and perinatal mortality and postnatal growth. Endocrinology 1997; 138: 2937-2947.
    • (1997) Endocrinology , vol.138 , pp. 2937-2947
    • Gay, E.1    Seurin, D.2    Babajko, S.3    Doublier, S.4    Cazillis, M.5    Binoux, M.6
  • 301
    • 0036714077 scopus 로고    scopus 로고
    • Hoxa5 overexpression correlates with IGFBP1 upregulation and postnatal dwarfism: Evidence for an interaction between Hoxa5 and Forkhead box transcription factors
    • Foucher I, Volovitch M, Frain M, et al. Hoxa5 overexpression correlates with IGFBP1 upregulation and postnatal dwarfism: evidence for an interaction between Hoxa5 and Forkhead box transcription factors. Development 2002; 129: 4065-4074.
    • (2002) Development , vol.129 , pp. 4065-4074
    • Foucher, I.1    Volovitch, M.2    Frain, M.3
  • 302
    • 34948872466 scopus 로고    scopus 로고
    • Transcriptional cross talk between the forkhead transcription factor forkhead box O1A and the progesterone receptor coordinates cell cycle regulation and differentiation in human endometrial stromal cells
    • Takano M, Lu Z, Goto T, et al. Transcriptional cross talk between the forkhead transcription factor forkhead box O1A and the progesterone receptor coordinates cell cycle regulation and differentiation in human endometrial stromal cells. Mol Endocrinol 2007; 21: 2334-2349.
    • (2007) Mol Endocrinol , vol.21 , pp. 2334-2349
    • Takano, M.1    Lu, Z.2    Goto, T.3
  • 303
    • 41549098778 scopus 로고    scopus 로고
    • The regulation and function of the forkhead transcription factor, Forkhead box O1, is dependent on the progesterone receptor in endometrial carcinoma
    • Ward EC, Hoekstra AV, Blok LJ, et al. The regulation and function of the forkhead transcription factor, Forkhead box O1, is dependent on the progesterone receptor in endometrial carcinoma. Endocrinology 2008; 149: 1942-1950.
    • (2008) Endocrinology , vol.149 , pp. 1942-1950
    • Ward, E.C.1    Hoekstra, A.V.2    Blok, L.J.3
  • 304
    • 0033546192 scopus 로고    scopus 로고
    • Phosphorylation of serine 256 by protein kinase B disrupts transactivation by FKHR and mediates effects of insulin on insulin¬like growth factor-binding protein-1 promoter activity through a conserved insulin response sequence
    • Guo S, Rena G, Cichy S, He X, Cohen P, Unterman T. Phosphorylation of serine 256 by protein kinase B disrupts transactivation by FKHR and mediates effects of insulin on insulin¬like growth factor-binding protein-1 promoter activity through a conserved insulin response sequence. J Biol Chem 1999; 274: 17184-17192.
    • (1999) J Biol Chem , vol.274 , pp. 17184-17192
    • Guo, S.1    Rena, G.2    Cichy, S.3    He, X.4    Cohen, P.5    Unterman, T.6
  • 305
    • 25144495477 scopus 로고    scopus 로고
    • Role of FOXO1A in the regulation of insulin-like growth factor-binding protein-1 in human endometrial cells: Interaction with progesterone receptor
    • Kim JJ, Buzzio OL, Li S, Lu Z. Role of FOXO1A in the regulation of insulin-like growth factor-binding protein-1 in human endometrial cells: interaction with progesterone receptor. Biol Reprod 2005; 73: 833-839.
    • (2005) Biol Reprod , vol.73 , pp. 833-839
    • Kim, J.J.1    Buzzio, O.L.2    Li, S.3    Lu, Z.4
  • 306
    • 34347384383 scopus 로고    scopus 로고
    • Constitutively active FOXO1a and a DNA-binding domain mutant exhibit distinct co-regulatory functions to enhance progesterone receptor A activity
    • Rudd MD, Gonzalez-Robayna I, Hernandez-Gonzalez I, Weigel NL, Bingman WE, 3rd, Richards JS. Constitutively active FOXO1a and a DNA-binding domain mutant exhibit distinct co-regulatory functions to enhance progesterone receptor A activity. J Mol Endocrinol 2007; 38: 673-690.
    • (2007) J Mol Endocrinol , vol.38 , pp. 673-690
    • Rudd, M.D.1    Gonzalez-Robayna, I.2    Hernandez-Gonzalez, I.3    Weigel, N.L.4    Bingman, W.E.5    Richards, J.S.6
  • 307
    • 18844432308 scopus 로고    scopus 로고
    • Adiponectin and adiponectin receptors
    • Kadowaki T, Yamauchi T. Adiponectin and adiponectin receptors. Endocr Rev 2005; 26: 439-451.
    • (2005) Endocr Rev , vol.26 , pp. 439-451
    • Kadowaki, T.1    Yamauchi, T.2
  • 308
    • 17544382289 scopus 로고    scopus 로고
    • AdipoQ is a novel adipose-specific gene dysregulated in obesity
    • Hu E, Liang P, Spiegelman BM. AdipoQ is a novel adipose-specific gene dysregulated in obesity. J Biol Chem 1996; 271: 10697-10703.
    • (1996) J Biol Chem , vol.271 , pp. 10697-10703
    • Hu, E.1    Liang, P.2    Spiegelman, B.M.3
  • 309
    • 33845985335 scopus 로고    scopus 로고
    • SIRT1 regulates adiponectin gene expression through Foxo1-C/enhancer-binding protein alpha transcriptional complex
    • Qiao L, Shao J. SIRT1 regulates adiponectin gene expression through Foxo1-C/enhancer-binding protein alpha transcriptional complex. J Biol Chem 2006; 281: 39915-39924.
    • (2006) J Biol Chem , vol.281 , pp. 39915-39924
    • Qiao, L.1    Shao, J.2
  • 310
    • 70350497395 scopus 로고    scopus 로고
    • Hematopoietic stem cell expansion precedes the generation of committed myeloid leukemia-initiating cells in C/EBPalpha mutant AML
    • Bereshchenko O, Mancini E, Moore S, et al. Hematopoietic stem cell expansion precedes the generation of committed myeloid leukemia-initiating cells in C/EBPalpha mutant AML. Cancer Cell 2009; 16: 390-400.
    • (2009) Cancer Cell , vol.16 , pp. 390-400
    • Bereshchenko, O.1    Mancini, E.2    Moore, S.3
  • 311
    • 0035374460 scopus 로고    scopus 로고
    • Growth hormone regulates phosphorylation and function of CCAAT/enhancer-binding protein beta by modulating Akt and glycogen synthase kinase-3
    • Piwien-Pilipuk G, Van Mater D, Ross SE, MacDougald OA, Schwartz J. Growth hormone regulates phosphorylation and function of CCAAT/enhancer-binding protein beta by modulating Akt and glycogen synthase kinase-3. J Biol Chem 2001; 276: 19664-19671.
    • (2001) J Biol Chem , vol.276 , pp. 19664-19671
    • Piwien-Pilipuk, G.1    van Mater, D.2    Ross, S.E.3    Macdougald, O.A.4    Schwartz, J.5
  • 312
    • 14644425319 scopus 로고    scopus 로고
    • Mechanisms of BCR-ABL in the pathogenesis of chronic myelogenous leukaemia
    • Ren R. Mechanisms of BCR-ABL in the pathogenesis of chronic myelogenous leukaemia. Nat Rev Cancer 2005; 5: 172-183.
    • (2005) Nat Rev Cancer , vol.5 , pp. 172-183
    • Ren, R.1
  • 313
    • 33749524667 scopus 로고    scopus 로고
    • Dynamic modeling of imatinib-treated chronic myeloid leukemia: Functional insights and clinical implications
    • Roeder I, Horn M, Glauche I, Hochhaus A, Mueller MC, Loeffler M. Dynamic modeling of imatinib-treated chronic myeloid leukemia: functional insights and clinical implications. Nat Med 2006; 12: 1181-1184.
    • (2006) Nat Med , vol.12 , pp. 1181-1184
    • Roeder, I.1    Horn, M.2    Glauche, I.3    Hochhaus, A.4    Mueller, M.C.5    Loeffler, M.6
  • 314
    • 76249087423 scopus 로고    scopus 로고
    • TGF-beta-FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia
    • Naka K, Hoshii T, Muraguchi T, et al. TGF-beta-FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia. Nature; 463: 676-680.
    • Nature , vol.463 , pp. 676-680
    • Naka, K.1    Hoshii, T.2    Muraguchi, T.3
  • 315
    • 0242580872 scopus 로고    scopus 로고
    • Convergence of peroxisome proliferator-activated receptor gamma and Foxo1 signaling pathways
    • Dowell P, Otto TC, Adi S, Lane MD. Convergence of peroxisome proliferator-activated receptor gamma and Foxo1 signaling pathways. J Biol Chem 2003; 278: 45485-45491.
    • (2003) J Biol Chem , vol.278 , pp. 45485-45491
    • Dowell, P.1    Otto, T.C.2    Adi, S.3    Lane, M.D.4
  • 316
    • 0037216526 scopus 로고    scopus 로고
    • AKT-independent protection of prostate cancer cells from apoptosis mediated through complex formation between the androgen receptor and FKHR
    • Li P, Lee H, Guo S, Unterman TG, Jenster G, Bai W. AKT-independent protection of prostate cancer cells from apoptosis mediated through complex formation between the androgen receptor and FKHR. Mol Cell Biol 2003; 23: 104-118.
    • (2003) Mol Cell Biol , vol.23 , pp. 104-118
    • Li, P.1    Lee, H.2    Guo, S.3    Unterman, T.G.4    Jenster, G.5    Bai, W.6
  • 317
    • 0035823576 scopus 로고    scopus 로고
    • Ligand-dependent interaction of estrogen receptor-alpha with members of the forkhead transcription factor family
    • Schuur ER, Loktev AV, Sharma M, Sun Z, Roth RA, Weigel RJ. Ligand-dependent interaction of estrogen receptor-alpha with members of the forkhead transcription factor family. J Biol Chem 2001; 276: 33554-33560.
    • (2001) J Biol Chem , vol.276 , pp. 33554-33560
    • Schuur, E.R.1    Loktev, A.V.2    Sharma, M.3    Sun, Z.4    Roth, R.A.5    Weigel, R.J.6
  • 318
    • 66449089902 scopus 로고    scopus 로고
    • FOXO1 transrepresses peroxisome proliferator-activated receptor gamma transactivation, coordinating an insulin-induced feed-forward response in adipocytes
    • Fan W, Imamura T, Sonoda N, et al. FOXO1 transrepresses peroxisome proliferator-activated receptor gamma transactivation, coordinating an insulin-induced feed-forward response in adipocytes. J Biol Chem 2009; 284: 12188-12197.
    • (2009) J Biol Chem , vol.284 , pp. 12188-12197
    • Fan, W.1    Imamura, T.2    Sonoda, N.3
  • 319
    • 0042405041 scopus 로고    scopus 로고
    • Peroxisome proliferator-activated receptors (PPARS): Regulators of gene expression in heart and skeletal muscle
    • Gilde AJ, Van Bilsen M. Peroxisome proliferator-activated receptors (PPARS): regulators of gene expression in heart and skeletal muscle. Acta Physiol Scand 2003; 178: 425-434.
    • (2003) Acta Physiol Scand , vol.178 , pp. 425-434
    • Gilde, A.J.1    van Bilsen, M.2
  • 320
    • 33746017379 scopus 로고    scopus 로고
    • FOXO1 represses peroxisome proliferator-activated receptor-gamma1 and -gamma2 gene promoters in primary adipocytes. A novel paradigm to increase insulin sensitivity
    • Armoni M, Harel C, Karni S, et al. FOXO1 represses peroxisome proliferator-activated receptor-gamma1 and -gamma2 gene promoters in primary adipocytes. A novel paradigm to increase insulin sensitivity. J Biol Chem 2006; 281: 19881-19891.
    • (2006) J Biol Chem , vol.281 , pp. 19881-19891
    • Armoni, M.1    Harel, C.2    Karni, S.3
  • 321
    • 64049089450 scopus 로고    scopus 로고
    • SIRT2 suppresses adipocyte differentiation by deacetylating FOXO1 and enhancing FOXO1's repressive interaction with PPARgamma
    • Wang F, Tong Q. SIRT2 suppresses adipocyte differentiation by deacetylating FOXO1 and enhancing FOXO1's repressive interaction with PPARgamma. Mol Biol Cell 2009; 20: 801-808.
    • (2009) Mol Biol Cell , vol.20 , pp. 801-808
    • Wang, F.1    Tong, Q.2
  • 322
    • 33747047885 scopus 로고    scopus 로고
    • Targeting foxo1 in mice using antisense oligonucleotide improves hepatic and peripheral insulin action
    • Samuel VT, Choi CS, Phillips TG, et al. Targeting foxo1 in mice using antisense oligonucleotide improves hepatic and peripheral insulin action. Diabetes 2006; 55: 2042-2050.
    • (2006) Diabetes , vol.55 , pp. 2042-2050
    • Samuel, V.T.1    Choi, C.S.2    Phillips, T.G.3
  • 323
    • 66649115077 scopus 로고    scopus 로고
    • FoxO1 haploinsufficiency protects against high-fat diet-induced insulin resistance with enhanced peroxisome proliferator-activated receptor gamma activation in adipose tissue
    • Kim JJ, Li P, Huntley J, Chang JP, Arden KC, Olefsky JM. FoxO1 haploinsufficiency protects against high-fat diet-induced insulin resistance with enhanced peroxisome proliferator-activated receptor gamma activation in adipose tissue. Diabetes 2009; 58: 1275-1282.
    • (2009) Diabetes , vol.58 , pp. 1275-1282
    • Kim, J.J.1    Li, P.2    Huntley, J.3    Chang, J.P.4    Arden, K.C.5    Olefsky, J.M.6
  • 324
    • 0033593461 scopus 로고    scopus 로고
    • Signaling mechanisms that regulate glucose transport
    • Czech MP, Corvera S. Signaling mechanisms that regulate glucose transport. J Biol Chem 1999; 274: 1865-1868.
    • (1999) J Biol Chem , vol.274 , pp. 1865-1868
    • Czech, M.P.1    Corvera, S.2
  • 325
    • 33947378695 scopus 로고    scopus 로고
    • Transcriptional regulation of the GLUT4 gene: From PPAR-gamma and FOXO1 to FFA and inflammation
    • Armoni M, Harel C, Karnieli E. Transcriptional regulation of the GLUT4 gene: from PPAR-gamma and FOXO1 to FFA and inflammation. Trends Endocrinol Metab 2007; 18: 100-107.
    • (2007) Trends Endocrinol Metab , vol.18 , pp. 100-107
    • Armoni, M.1    Harel, C.2    Karnieli, E.3
  • 326
    • 0036846196 scopus 로고    scopus 로고
    • PAX3/forkhead homolog in rhabdomyosarcoma oncoprotein activates glucose transporter 4 gene expression in vivo and in vitro
    • Armoni M, Quon MJ, Maor G, et al. PAX3/forkhead homolog in rhabdomyosarcoma oncoprotein activates glucose transporter 4 gene expression in vivo and in vitro. J Clin Endocrinol Metab 2002; 87: 5312-5324.
    • (2002) J Clin Endocrinol Metab , vol.87 , pp. 5312-5324
    • Armoni, M.1    Quon, M.J.2    Maor, G.3
  • 327
    • 0037432191 scopus 로고    scopus 로고
    • A forkhead transcription factor FKHR up-regulates lipoprotein lipase expression in skeletal muscle
    • Kamei Y, Mizukami J, Miura S, et al. A forkhead transcription factor FKHR up-regulates lipoprotein lipase expression in skeletal muscle. FEBS Lett 2003; 536: 232-236.
    • (2003) FEBS Lett , vol.536 , pp. 232-236
    • Kamei, Y.1    Mizukami, J.2    Miura, S.3
  • 328
    • 17144428529 scopus 로고    scopus 로고
    • FoxO1 stimulates fatty acid uptake and oxidation in muscle cells through CD36-dependent and -independent mechanisms
    • Bastie CC, Nahle Z, McLoughlin T, et al. FoxO1 stimulates fatty acid uptake and oxidation in muscle cells through CD36-dependent and -independent mechanisms. J Biol Chem 2005; 280: 14222-14229.
    • (2005) J Biol Chem , vol.280 , pp. 14222-14229
    • Bastie, C.C.1    Nahle, Z.2    McLoughlin, T.3
  • 329
    • 0035139016 scopus 로고    scopus 로고
    • The role of PPAR-gamma in macrophage differentiation and cholesterol uptake
    • Moore KJ, Rosen ED, Fitzgerald ML, et al. The role of PPAR-gamma in macrophage differentiation and cholesterol uptake. Nat Med 2001; 7: 41-47.
    • (2001) Nat Med , vol.7 , pp. 41-47
    • Moore, K.J.1    Rosen, E.D.2    Fitzgerald, M.L.3
  • 330
    • 0035132330 scopus 로고    scopus 로고
    • PPAR-gamma dependent and independent effects on macrophage-gene expression in lipid metabolism and inflammation
    • Chawla A, Barak Y, Nagy L, Liao D, Tontonoz P, Evans RM. PPAR-gamma dependent and independent effects on macrophage-gene expression in lipid metabolism and inflammation. Nat Med 2001; 7: 48-52.
    • (2001) Nat Med , vol.7 , pp. 48-52
    • Chawla, A.1    Barak, Y.2    Nagy, L.3    Liao, D.4    Tontonoz, P.5    Evans, R.M.6
  • 331
    • 33846878069 scopus 로고    scopus 로고
    • PPAR{alpha} mediates the hypolipidemic action of fibrates by antagonizing FoxO1
    • Qu S, Su D, Altomonte J, et al. PPAR{alpha} mediates the hypolipidemic action of fibrates by antagonizing FoxO1. Am J Physiol Endocrinol Metab 2007; 292: E421-E434.
    • (2007) Am J Physiol Endocrinol Metab , vol.292 , pp. 421-434
    • Qu, S.1    Su, D.2    Altomonte, J.3
  • 332
    • 75149152919 scopus 로고    scopus 로고
    • Metabolism of Very-Low-Density Lipoprotein and Low-Density Lipoprotein Containing Apolipoprotein C-III and Not Other Small Apolipoproteins
    • Mendivil CO, Zheng C, Furtado J, Lel J, Sacks FM. Metabolism of Very-Low-Density Lipoprotein and Low-Density Lipoprotein Containing Apolipoprotein C-III and Not Other Small Apolipoproteins. Arterioscler Thromb Vasc Biol 2010; 30: 239-245.
    • (2010) Arterioscler Thromb Vasc Biol , vol.30 , pp. 239-245
    • Mendivil, C.O.1    Zheng, C.2    Furtado, J.3    Lel, J.4    Sacks, F.M.5
  • 333
    • 15244339047 scopus 로고    scopus 로고
    • Foxo1 mediates insulin action on apoC-III and triglyceride metabolism
    • Altomonte J, Cong L, Harbaran S, et al. Foxo1 mediates insulin action on apoC-III and triglyceride metabolism. J Clin Invest 2004; 114: 1493-1503.
    • (2004) J Clin Invest , vol.114 , pp. 1493-1503
    • Altomonte, J.1    Cong, L.2    Harbaran, S.3
  • 334
    • 0038242819 scopus 로고    scopus 로고
    • TRB3: A tribbles homolog that inhibits Akt/PKB activation by insulin in liver
    • Du K, Herzig S, Kulkarni RN, Montminy M. TRB3: a tribbles homolog that inhibits Akt/PKB activation by insulin in liver. Science 2003; 300: 1574-1577.
    • (2003) Science , vol.300 , pp. 1574-1577
    • Du, K.1    Herzig, S.2    Kulkarni, R.N.3    Montminy, M.4
  • 335
    • 2442701392 scopus 로고    scopus 로고
    • PGC-1 promotes insulin resistance in liver through PPAR-alpha-dependent induction of TRB-3
    • Koo SH, Satoh H, Herzig S, et al. PGC-1 promotes insulin resistance in liver through PPAR-alpha-dependent induction of TRB-3. Nat Med 2004; 10: 530-534.
    • (2004) Nat Med , vol.10 , pp. 530-534
    • Koo, S.H.1    Satoh, H.2    Herzig, S.3
  • 336
    • 34547956322 scopus 로고    scopus 로고
    • Three members of the human pyruvate dehydrogenase kinase gene family are direct targets of the peroxisome proliferator-activated receptor beta/delta
    • Degenhardt T, Saramaki A, Malinen M, et al. Three members of the human pyruvate dehydrogenase kinase gene family are direct targets of the peroxisome proliferator-activated receptor beta/delta. J Mol Biol 2007; 372: 341-355.
    • (2007) J Mol Biol , vol.372 , pp. 341-355
    • Degenhardt, T.1    Saramaki, A.2    Malinen, M.3
  • 337
    • 0037135623 scopus 로고    scopus 로고
    • Fatty acid homeostasis and induction of lipid regulatory genes in skeletal muscles of peroxisome proliferator-activated receptor (PPAR) alpha knock-out mice. Evidence for compensatory regulation by PPAR delta
    • Muoio DM, MacLean PS, Lang DB, et al. Fatty acid homeostasis and induction of lipid regulatory genes in skeletal muscles of peroxisome proliferator-activated receptor (PPAR) alpha knock-out mice. Evidence for compensatory regulation by PPAR delta. J Biol Chem 2002; 277: 26089-26097.
    • (2002) J Biol Chem , vol.277 , pp. 26089-26097
    • Muoio, D.M.1    Maclean, P.S.2    Lang, D.B.3
  • 338
    • 34547673326 scopus 로고    scopus 로고
    • Regulatory functions of PPARbeta in metabolism: Implications for the treatment of metabolic syndrome
    • Grimaldi PA. Regulatory functions of PPARbeta in metabolism: implications for the treatment of metabolic syndrome. Biochim Biophys Acta 2007; 1771: 983-990.
    • (2007) Biochim Biophys Acta , vol.1771 , pp. 983-990
    • Grimaldi, P.A.1
  • 339
    • 33646792244 scopus 로고    scopus 로고
    • Peroxisome proliferator-activated receptor delta (PPARdelta), a novel target site for drug discovery in metabolic syndrome
    • Takahashi S, Tanaka T, Kodama T, Sakai J. Peroxisome proliferator-activated receptor delta (PPARdelta), a novel target site for drug discovery in metabolic syndrome. Pharmacol Res 2006; 53: 501-507.
    • (2006) Pharmacol Res , vol.53 , pp. 501-507
    • Takahashi, S.1    Tanaka, T.2    Kodama, T.3    Sakai, J.4
  • 340
    • 47249119658 scopus 로고    scopus 로고
    • CD36-dependent regulation of muscle FoxO1 and PDK4 in the PPAR delta/beta-mediated adaptation to metabolic stress
    • Nahle Z, Hsieh M, Pietka T, et al. CD36-dependent regulation of muscle FoxO1 and PDK4 in the PPAR delta/beta-mediated adaptation to metabolic stress. J Biol Chem 2008; 283: 14317-14326.
    • (2008) J Biol Chem , vol.283 , pp. 14317-14326
    • Nahle, Z.1    Hsieh, M.2    Pietka, T.3
  • 341
    • 52649174345 scopus 로고    scopus 로고
    • Pharmacological activation of PPARbeta promotes rapid and calcineurin-dependent fiber remodeling and angiogenesis in mouse skeletal muscle
    • Gaudel C, Schwartz C, Giordano C, Abumrad NA, Grimaldi PA. Pharmacological activation of PPARbeta promotes rapid and calcineurin-dependent fiber remodeling and angiogenesis in mouse skeletal muscle. Am J Physiol Endocrinol Metab 2008; 295: E297-E304.
    • (2008) Am J Physiol Endocrinol Metab , vol.295 , pp. 297-304
    • Gaudel, C.1    Schwartz, C.2    Giordano, C.3    Abumrad, N.A.4    Grimaldi, P.A.5
  • 342
    • 34547865385 scopus 로고    scopus 로고
    • PPARdelta agonism induces a change in fuel metabolism and activation of an atrophy programme, but does not impair mitochondrial function in rat skeletal muscle
    • Constantin D, Constantin-Teodosiu D, Layfield R, Tsintzas K, Bennett AJ, Greenhaff PL. PPARdelta agonism induces a change in fuel metabolism and activation of an atrophy programme, but does not impair mitochondrial function in rat skeletal muscle. J Physiol 2007; 583: 381-390.
    • (2007) J Physiol , vol.583 , pp. 381-390
    • Constantin, D.1    Constantin-Teodosiu, D.2    Layfield, R.3    Tsintzas, K.4    Bennett, A.J.5    Greenhaff, P.L.6
  • 343
    • 0034518420 scopus 로고    scopus 로고
    • The role of orphan nuclear receptors in the regulation of cholesterol homeostasis
    • Repa JJ, Mangelsdorf DJ. The role of orphan nuclear receptors in the regulation of cholesterol homeostasis. Annu Rev Cell Dev Biol 2000; 16: 459-481.
    • (2000) Annu Rev Cell Dev Biol , vol.16 , pp. 459-481
    • Repa, J.J.1    Mangelsdorf, D.J.2
  • 344
    • 0013199471 scopus 로고    scopus 로고
    • Cholesterol and bile acid metabolism are impaired in mice lacking the nuclear oxysterol receptor LXR alpha
    • Peet DJ, Turley SD, Ma W, et al. Cholesterol and bile acid metabolism are impaired in mice lacking the nuclear oxysterol receptor LXR alpha. Cell 1998; 93: 693-704.
    • (1998) Cell , vol.93 , pp. 693-704
    • Peet, D.J.1    Turley, S.D.2    Ma, W.3
  • 345
    • 0037058995 scopus 로고    scopus 로고
    • Disruption of Abcg5 and Abcg8 in mice reveals their crucial role in biliary cholesterol secretion
    • Yu L, Hammer RE, Li-Hawkins J, et al. Disruption of Abcg5 and Abcg8 in mice reveals their crucial role in biliary cholesterol secretion. Proc Natl Acad Sci USA 2002; 99: 16237-16242.
    • (2002) Proc Natl Acad Sci USA , vol.99 , pp. 16237-16242
    • Yu, L.1    Hammer, R.E.2    Li-Hawkins, J.3
  • 346
    • 34547652884 scopus 로고    scopus 로고
    • Ligand activation of LXR beta reverses atherosclerosis and cellular cholesterol overload in mice lacking LXR alpha and apoE
    • Bradley MN, Hong C, Chen M, et al. Ligand activation of LXR beta reverses atherosclerosis and cellular cholesterol overload in mice lacking LXR alpha and apoE. J Clin Invest 2007; 117: 2337-2346.
    • (2007) J Clin Invest , vol.117 , pp. 2337-2346
    • Bradley, M.N.1    Hong, C.2    Chen, M.3
  • 347
    • 42549086338 scopus 로고    scopus 로고
    • Regulation of SREBP1c gene expression in skeletal muscle: Role of retinoid X receptor/liver X receptor and forkhead-O1 transcription factor
    • Kamei Y, Miura S, Suganami T, et al. Regulation of SREBP1c gene expression in skeletal muscle: role of retinoid X receptor/liver X receptor and forkhead-O1 transcription factor. Endocrinology 2008; 149: 2293-2305.
    • (2008) Endocrinology , vol.149 , pp. 2293-2305
    • Kamei, Y.1    Miura, S.2    Suganami, T.3
  • 348
    • 42249111748 scopus 로고    scopus 로고
    • The roles of nuclear receptors CAR and PXR in hepatic energy metabolism
    • Konno Y, Negishi M, Kodama S. The roles of nuclear receptors CAR and PXR in hepatic energy metabolism. Drug Metab Pharmacokinet 2008; 23: 8-13.
    • (2008) Drug Metab Pharmacokinet , vol.23 , pp. 8-13
    • Konno, Y.1    Negishi, M.2    Kodama, S.3
  • 350
    • 33744948052 scopus 로고    scopus 로고
    • A novel pregnane X receptor-mediated and sterol regulatory element-binding protein-independent lipogenic pathway
    • Zhou J, Zhai Y, Mu Y, et al. A novel pregnane X receptor-mediated and sterol regulatory element-binding protein-independent lipogenic pathway. J Biol Chem 2006; 281: 15013-15020.
    • (2006) J Biol Chem , vol.281 , pp. 15013-15020
    • Zhou, J.1    Zhai, Y.2    Mu, Y.3
  • 351
    • 35748972683 scopus 로고    scopus 로고
    • Human nuclear pregnane X receptor cross-talk with CREB to repress cAMP activation of the glucose-6-phosphatase gene
    • Kodama S, Moore R, Yamamoto Y, Negishi M. Human nuclear pregnane X receptor cross-talk with CREB to repress cAMP activation of the glucose-6-phosphatase gene. Biochem J 2007; 407: 373-381.
    • (2007) Biochem J , vol.407 , pp. 373-381
    • Kodama, S.1    Moore, R.2    Yamamoto, Y.3    Negishi, M.4
  • 352
    • 4444246000 scopus 로고    scopus 로고
    • Nuclear receptors CAR and PXR cross talk with FOXO1 to regulate genes that encode drug-metabolizing and gluconeogenic enzymes
    • Kodama S, Koike C, Negishi M, Yamamoto Y. Nuclear receptors CAR and PXR cross talk with FOXO1 to regulate genes that encode drug-metabolizing and gluconeogenic enzymes. Mol Cell Biol 2004; 24: 7931-7940.
    • (2004) Mol Cell Biol , vol.24 , pp. 7931-7940
    • Kodama, S.1    Koike, C.2    Negishi, M.3    Yamamoto, Y.4
  • 353
    • 33847240106 scopus 로고    scopus 로고
    • (Far) Outside the box: Genomic approach to acute porphyria
    • Thunell S. (Far) Outside the box: genomic approach to acute porphyria. Physiol Res 2006; 55: S43-S66.
    • (2006) Physiol Res , vol.55 , pp. 43-66
    • Thunell, S.1
  • 354
    • 0029562554 scopus 로고
    • The RXR heterodimers and orphan receptors
    • Mangelsdorf DJ, Evans RM. The RXR heterodimers and orphan receptors. Cell 1995; 83: 841-850.
    • (1995) Cell , vol.83 , pp. 841-850
    • Mangelsdorf, D.J.1    Evans, R.M.2
  • 355
    • 27844497945 scopus 로고    scopus 로고
    • FOXO transcription factors at the interface between longevity and tumor suppression
    • Greer EL, Brunet A. FOXO transcription factors at the interface between longevity and tumor suppression. Oncogene 2005; 24: 7410-7425.
    • (2005) Oncogene , vol.24 , pp. 7410-7425
    • Greer, E.L.1    Brunet, A.2
  • 356
    • 33646782964 scopus 로고    scopus 로고
    • FOXO factors: A matter of life and death
    • Huang H, Tindall DJ. FOXO factors: a matter of life and death. Future Oncol 2006; 2: 83-89.
    • (2006) Future Oncol , vol.2 , pp. 83-89
    • Huang, H.1    Tindall, D.J.2
  • 357
    • 25444496757 scopus 로고    scopus 로고
    • Chemistry and structural biology of androgen receptor
    • Gao W, Bohl CE, Dalton JT. Chemistry and structural biology of androgen receptor. Chem Rev 2005; 105: 3352-3370.
    • (2005) Chem Rev , vol.105 , pp. 3352-3370
    • Gao, W.1    Bohl, C.E.2    Dalton, J.T.3
  • 358
    • 0035912833 scopus 로고    scopus 로고
    • Akt suppresses androgen-induced apoptosis by phosphorylating and inhibiting androgen receptor
    • Lin HK, Yeh S, Kang HY, Chang C. Akt suppresses androgen-induced apoptosis by phosphorylating and inhibiting androgen receptor. Proc Natl Acad Sci USA 2001; 98: 7200-7205.
    • (2001) Proc Natl Acad Sci USA , vol.98 , pp. 7200-7205
    • Lin, H.K.1    Yeh, S.2    Kang, H.Y.3    Chang, C.4
  • 359
    • 0035827510 scopus 로고    scopus 로고
    • Antagonism between PTEN/MMAC1/TEP-1 and androgen receptor in growth and apoptosis of prostatic cancer cells
    • Li P, Nicosia SV, Bai W. Antagonism between PTEN/MMAC1/TEP-1 and androgen receptor in growth and apoptosis of prostatic cancer cells. J Biol Chem 2001; 276: 20444-20450.
    • (2001) J Biol Chem , vol.276 , pp. 20444-20450
    • Li, P.1    Nicosia, S.V.2    Bai, W.3
  • 360
    • 33746930170 scopus 로고    scopus 로고
    • FOXO1A is a candidate for the 13q14 tumor suppressor gene inhibiting androgen receptor signaling in prostate cancer
    • Dong XY, Chen C, Sun X, et al. FOXO1A is a candidate for the 13q14 tumor suppressor gene inhibiting androgen receptor signaling in prostate cancer. Cancer Res 2006; 66: 6998-7006.
    • (2006) Cancer Res , vol.66 , pp. 6998-7006
    • Dong, X.Y.1    Chen, C.2    Sun, X.3
  • 361
    • 0030669765 scopus 로고    scopus 로고
    • Interaction between the amino-and carboxyl-terminal regions of the rat androgen receptor modulates transcriptional activity and is influenced by nuclear receptor coactivators
    • Ikonen T, Palvimo JJ, Janne OA. Interaction between the amino-and carboxyl-terminal regions of the rat androgen receptor modulates transcriptional activity and is influenced by nuclear receptor coactivators. J Biol Chem. 1997; 272: 29821-29828.
    • (1997) J Biol Chem , vol.272 , pp. 29821-29828
    • Ikonen, T.1    Palvimo, J.J.2    Janne, O.A.3
  • 362
    • 0029560494 scopus 로고
    • Evidence for an anti-parallel orientation of the ligand-activated human androgen receptor dimer
    • Langley E, Zhou ZX, Wilson EM. Evidence for an anti-parallel orientation of the ligand-activated human androgen receptor dimer. J Biol Chem 1995; 270: 29983-29990.
    • (1995) J Biol Chem , vol.270 , pp. 29983-29990
    • Langley, E.1    Zhou, Z.X.2    Wilson, E.M.3
  • 363
    • 58849127943 scopus 로고    scopus 로고
    • FoxO1 mediates PTEN suppression of androgen receptor N- and C-terminal interactions and coactivator recruitment
    • Ma Q, Fu W, Li P, et al. FoxO1 mediates PTEN suppression of androgen receptor N- and C-terminal interactions and coactivator recruitment. Mol Endocrinol 2009; 23: 213-225.
    • (2009) Mol Endocrinol , vol.23 , pp. 213-225
    • Ma, Q.1    Fu, W.2    Li, P.3
  • 364
    • 34147101043 scopus 로고    scopus 로고
    • Insulin-like growth factor 1/insulin signaling activates androgen signaling through direct interactions of Foxo1 with androgen receptor
    • Fan W, Yanase T, Morinaga H, et al. Insulin-like growth factor 1/insulin signaling activates androgen signaling through direct interactions of Foxo1 with androgen receptor. J Biol Chem 2007; 282: 7329-7338.
    • (2007) J Biol Chem , vol.282 , pp. 7329-7338
    • Fan, W.1    Yanase, T.2    Morinaga, H.3
  • 365
    • 47949126042 scopus 로고    scopus 로고
    • FOXO3a mediates the androgen-dependent regulation of FLIP and contributes to TRAIL-induced apoptosis of LNCaP cells
    • Cornforth AN, Davis JS, Khanifar E, Nastiuk KL, Krolewski JJ. FOXO3a mediates the androgen-dependent regulation of FLIP and contributes to TRAIL-induced apoptosis of LNCaP cells. Oncogene 2008; 27: 4422-4433.
    • (2008) Oncogene , vol.27 , pp. 4422-4433
    • Cornforth, A.N.1    Davis, J.S.2    Khanifar, E.3    Nastiuk, K.L.4    Krolewski, J.J.5
  • 366
    • 61749091464 scopus 로고    scopus 로고
    • Modification of androgen receptor function by IGF-1 signaling implications in the mechanism of refractory prostate carcinoma
    • Yanase T, Fan W. Modification of androgen receptor function by IGF-1 signaling implications in the mechanism of refractory prostate carcinoma. Vitam Horm 2009; 80: 649-666.
    • (2009) Vitam Horm , vol.80 , pp. 649-666
    • Yanase, T.1    Fan, W.2
  • 367
    • 33644638521 scopus 로고    scopus 로고
    • Estrogen receptors and human disease
    • Deroo BJ, Korach KS. Estrogen receptors and human disease. J Clin Invest 2006; 116: 561-570.
    • (2006) J Clin Invest , vol.116 , pp. 561-570
    • Deroo, B.J.1    Korach, K.S.2
  • 368
    • 0031240327 scopus 로고    scopus 로고
    • Mammary gland development and tumorigenesis in estrogen receptor knockout mice
    • Bocchinfuso WP, Korach KS. Mammary gland development and tumorigenesis in estrogen receptor knockout mice. J Mammary Gland Biol Neoplasia 1997; 2: 323-334.
    • (1997) J Mammary Gland Biol Neoplasia , vol.2 , pp. 323-334
    • Bocchinfuso, W.P.1    Korach, K.S.2
  • 369
    • 31544451511 scopus 로고    scopus 로고
    • RNA interference-mediated depletion of phosphoinositide 3-kinase activates forkhead box class O transcription factors and induces cell cycle arrest and apoptosis in breast carcinoma cells
    • Reagan-Shaw S, Ahmad N. RNA interference-mediated depletion of phosphoinositide 3-kinase activates forkhead box class O transcription factors and induces cell cycle arrest and apoptosis in breast carcinoma cells. Cancer Res 2006; 66: 1062-1069.
    • (2006) Cancer Res , vol.66 , pp. 1062-1069
    • Reagan-Shaw, S.1    Ahmad, N.2
  • 370
    • 10744230034 scopus 로고    scopus 로고
    • FoxO3a transcriptional regulation of Bim controls apoptosis in paclitaxel-treated breast cancer cell lines
    • Sunters A, Fernandez de Mattos S, Stahl M, et al. FoxO3a transcriptional regulation of Bim controls apoptosis in paclitaxel-treated breast cancer cell lines. J Biol Chem 2003; 278: 49795-49805.
    • (2003) J Biol Chem , vol.278 , pp. 49795-49805
    • Sunters, A.1    de Mattos, F.S.2    Stahl, M.3
  • 371
    • 0037472647 scopus 로고    scopus 로고
    • Estrogen regulation of Pak1 and FKHR pathways in breast cancer cells
    • azumdar A, Kumar R. Estrogen regulation of Pak1 and FKHR pathways in breast cancer cells. FEBS Lett 2003; 535: 6-10.
    • (2003) FEBS Lett , vol.535 , pp. 6-10
    • Azumdar, A.1    Kumar, R.2
  • 372
    • 44849127978 scopus 로고    scopus 로고
    • Forkhead box transcription factor FOXO3a suppresses estrogen-dependent breast cancer cell proliferation and tumorigenesis
    • Zou Y, Tsai WB, Cheng CJ, et al. Forkhead box transcription factor FOXO3a suppresses estrogen-dependent breast cancer cell proliferation and tumorigenesis. Breast Cancer Res 2008; 10: R21.
    • (2008) Breast Cancer Res , vol.10
    • Zou, Y.1    Tsai, W.B.2    Cheng, C.J.3
  • 373
    • 22844438667 scopus 로고    scopus 로고
    • Crosstalk between p53 and FOXO transcription factors
    • You H, Mak TW. Crosstalk between p53 and FOXO transcription factors. Cell Cycle 2005; 4: 37-38.
    • (2005) Cell Cycle , vol.4 , pp. 37-38
    • You, H.1    Mak, T.W.2
  • 374
    • 70350565471 scopus 로고    scopus 로고
    • Evan GI. p53--a Jack of all trades but master of none
    • Junttila MR, Evan GI. p53--a Jack of all trades but master of none. Nat Rev Cancer 2009; 9: 821-829.
    • (2009) Nat Rev Cancer , vol.9 , pp. 821-829
    • Junttila, M.R.1
  • 375
    • 33745163907 scopus 로고    scopus 로고
    • Regulation of transactivation-independent proapoptotic activity of p53 by FOXO3a
    • You H, Yamamoto K, Mak TW. Regulation of transactivation-independent proapoptotic activity of p53 by FOXO3a. Proc Natl Acad Sci USA 2006; 103: 9051-9056.
    • (2006) Proc Natl Acad Sci USA , vol.103 , pp. 9051-9056
    • You, H.1    Yamamoto, K.2    Mak, T.W.3
  • 376
    • 10844236451 scopus 로고    scopus 로고
    • Nutrient availability regulates SIRT1 through a forkhead-dependent pathway
    • Nemoto S, Fergusson MM, Finkel T. Nutrient availability regulates SIRT1 through a forkhead-dependent pathway. Science 2004; 306: 2105-2108.
    • (2004) Science , vol.306 , pp. 2105-2108
    • Nemoto, S.1    Fergusson, M.M.2    Finkel, T.3
  • 377
    • 0037349289 scopus 로고    scopus 로고
    • P53 has a direct apoptogenic role at the mitochondria
    • Mihara M, Erster S, Zaika A, et al. P53 has a direct apoptogenic role at the mitochondria. Mol Cell 2003; 11: 577-590.
    • (2003) Mol Cell , vol.11 , pp. 577-590
    • Mihara, M.1    Erster, S.2    Zaika, A.3
  • 378
    • 68049111575 scopus 로고    scopus 로고
    • P53 negatively regulates the transcriptional activity of FOXO3a under oxidative stress
    • Miyaguchi Y, Tsuchiya K, Sakamoto K. P53 negatively regulates the transcriptional activity of FOXO3a under oxidative stress. Cell Biol Int 2009; 33: 853-860.
    • (2009) Cell Biol Int , vol.33 , pp. 853-860
    • Miyaguchi, Y.1    Tsuchiya, K.2    Sakamoto, K.3
  • 379
    • 33846214693 scopus 로고    scopus 로고
    • Regulation of the Arf tumor suppressor in Emicro-Myc transgenic mice: Longitudinal study of Myc-induced lymphomagenesis
    • Bertwistle D, Sherr CJ. Regulation of the Arf tumor suppressor in Emicro-Myc transgenic mice: longitudinal study of Myc-induced lymphomagenesis. Blood 2007; 109: 792-794.
    • (2007) Blood , vol.109 , pp. 792-794
    • Bertwistle, D.1    Sherr, C.J.2
  • 380
    • 13144253122 scopus 로고    scopus 로고
    • Reversing drug resistance in vivo
    • Wendel HG, Lowe SW. Reversing drug resistance in vivo. Cell Cycle 2004; 3: 847-849.
    • (2004) Cell Cycle , vol.3 , pp. 847-849
    • Wendel, H.G.1    Lowe, S.W.2
  • 381
    • 3543028648 scopus 로고    scopus 로고
    • Myc-induced proliferation and transformation require Akt-mediated phosphorylation of FoxO proteins
    • Bouchard C, Marquardt J, Bras A, Medema RH, Eilers M. Myc-induced proliferation and transformation require Akt-mediated phosphorylation of FoxO proteins. Embo J 2004; 23:2830-2840.
    • (2004) Embo J , vol.23 , pp. 2830-2840
    • Bouchard, C.1    Marquardt, J.2    Bras, A.3    Medema, R.H.4    Eilers, M.5
  • 382
    • 0020520516 scopus 로고
    • Tumorigenic conversion of primary embryo fibroblasts requires at least two cooperating oncogenes
    • Land H, Parada LF, Weinberg RA. Tumorigenic conversion of primary embryo fibroblasts requires at least two cooperating oncogenes. Nature 1983; 304: 596-602.
    • (1983) Nature , vol.304 , pp. 596-602
    • Land, H.1    Parada, L.F.2    Weinberg, R.A.3
  • 383
    • 35948946514 scopus 로고    scopus 로고
    • FoxO transcription factors suppress Myc-driven lymphomagenesis via direct activation of Arf
    • Bouchard C, Lee S, Paulus-Hock V, Loddenkemper C, Eilers M, Schmitt CA. FoxO transcription factors suppress Myc-driven lymphomagenesis via direct activation of Arf. Genes Dev 2007; 21: 2775-2787.
    • (2007) Genes Dev , vol.21 , pp. 2775-2787
    • Bouchard, C.1    Lee, S.2    Paulus-Hock, V.3    Loddenkemper, C.4    Eilers, M.5    Schmitt, C.A.6
  • 384
    • 2142662252 scopus 로고    scopus 로고
    • Reciprocal control of Forkhead box O 3a and c-Myc via the phosphatidylinositol 3-kinase pathway coordinately regulates p27Kip1 levels
    • Chandramohan V, Jeay S, Pianetti S, Sonenshein GE. Reciprocal control of Forkhead box O 3a and c-Myc via the phosphatidylinositol 3-kinase pathway coordinately regulates p27Kip1 levels. J Immunol 2004; 172: 5522-5527.
    • (2004) J Immunol , vol.172 , pp. 5522-5527
    • Chandramohan, V.1    Jeay, S.2    Pianetti, S.3    Sonenshein, G.E.4
  • 385
    • 0035967099 scopus 로고    scopus 로고
    • Repression of transcription of the p27(Kip1) cyclin-dependent kinase inhibitor gene by c-Myc
    • Yang W, Shen J, Wu M, et al. Repression of transcription of the p27(Kip1) cyclin-dependent kinase inhibitor gene by c-Myc. Oncogene 2001; 20: 1688-1702.
    • (2001) Oncogene , vol.20 , pp. 1688-1702
    • Yang, W.1    Shen, J.2    Wu, M.3
  • 386
    • 49049100243 scopus 로고    scopus 로고
    • C-Myc represses FOXO3a-mediated transcription of the gene encoding the p27(Kip1) cyclin dependent kinase inhibitor
    • Chandramohan V, Mineva ND, Burke B, et al. C-Myc represses FOXO3a-mediated transcription of the gene encoding the p27(Kip1) cyclin dependent kinase inhibitor. J Cell Biochem 2008; 104: 2091-2106.
    • (2008) J Cell Biochem , vol.104 , pp. 2091-2106
    • Chandramohan, V.1    Mineva, N.D.2    Burke, B.3
  • 387
    • 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 Jr., J.E.3
  • 388
    • 0032525155 scopus 로고    scopus 로고
    • Multiple routes to astrocytic differentiation in the CNS
    • Rajan P, McKay RD. Multiple routes to astrocytic differentiation in the CNS. J Neurosci 1998; 18: 3620-3629.
    • (1998) J Neurosci , vol.18 , pp. 3620-3629
    • Rajan, P.1    McKay, R.D.2
  • 389
    • 33746397211 scopus 로고    scopus 로고
    • Non-cell-autonomous action of STAT3 in maintenance of neural precursor cells in the mouse neocortex
    • Yoshimatsu T, Kawaguchi D, Oishi K, et al. Non-cell-autonomous action of STAT3 in maintenance of neural precursor cells in the mouse neocortex. Development 2006; 133: 2553-2563.
    • (2006) Development , vol.133 , pp. 2553-2563
    • Yoshimatsu, T.1    Kawaguchi, D.2    Oishi, K.3
  • 390
    • 39449093328 scopus 로고    scopus 로고
    • Identification of a PTEN-regulated STAT3 brain tumor suppressor pathway
    • de la Iglesia N, Konopka G, Puram SV, et al. Identification of a PTEN-regulated STAT3 brain tumor suppressor pathway. Genes Dev 2008; 22: 449-462.
    • (2008) Genes Dev , vol.22 , pp. 449-462
    • de la Iglesia, N.1    Konopka, G.2    Puram, S.V.3
  • 391
    • 2442657786 scopus 로고    scopus 로고
    • IL-6 is required for glioma development in a mouse model
    • Weissenberger J, Loeffler S, Kappeler A, et al. IL-6 is required for glioma development in a mouse model. Oncogene 2004; 23: 3308-3316.
    • (2004) Oncogene , vol.23 , pp. 3308-3316
    • Weissenberger, J.1    Loeffler, S.2    Kappeler, A.3
  • 392
    • 25444454543 scopus 로고    scopus 로고
    • New and old functions of STAT3: A pivotal target for individualized treatment of cancer
    • Inghirami G, Chiarle R, Simmons WJ, Piva R, Schlessinger K, Levy DE. New and old functions of STAT3: a pivotal target for individualized treatment of cancer. Cell Cycle 2005; 4: 1131-1133.
    • (2005) Cell Cycle , vol.4 , pp. 1131-1133
    • Inghirami, G.1    Chiarle, R.2    Simmons, W.J.3    Piva, R.4    Schlessinger, K.5    Levy, D.E.6
  • 393
    • 3042748279 scopus 로고    scopus 로고
    • Knockdown of STAT3 expression by RNAi induces apoptosis in astrocytoma cells
    • Konnikova L, Kotecki M, Kruger MM, Cochran BH. Knockdown of STAT3 expression by RNAi induces apoptosis in astrocytoma cells. BMC Cancer 2003; 3: 23.
    • (2003) BMC Cancer , vol.3 , pp. 23
    • Konnikova, L.1    Kotecki, M.2    Kruger, M.M.3    Cochran, B.H.4
  • 394
    • 61449095573 scopus 로고    scopus 로고
    • Leptin at 14 y of age: An ongoing story
    • Friedman JM. Leptin at 14 y of age: an ongoing story. Am J Clin Nutr 2009; 89: 973S-979S.
    • (2009) Am J Clin Nutr , vol.89
    • Friedman, J.M.1
  • 395
    • 17844401704 scopus 로고    scopus 로고
    • Anatomy and regulation of the central melanocortin system
    • Cone RD. Anatomy and regulation of the central melanocortin system. Nat Neurosci 2005; 8: 571-578.
    • (2005) Nat Neurosci , vol.8 , pp. 571-578
    • Cone, R.D.1
  • 396
    • 0037456304 scopus 로고    scopus 로고
    • STAT3 signalling is required for leptin regulation of energy balance but not reproduction
    • Bates SH, Stearns WH, Dundon TA, et al. STAT3 signalling is required for leptin regulation of energy balance but not reproduction. Nature 2003; 421: 856-859.
    • (2003) Nature , vol.421 , pp. 856-859
    • Bates, S.H.1    Stearns, W.H.2    Dundon, T.A.3
  • 397
    • 11144357516 scopus 로고    scopus 로고
    • Role of STAT-3 in regulation of hepatic gluconeogenic genes and carbohydrate metabolism in vivo
    • Inoue H, Ogawa W, Ozaki M, et al. Role of STAT-3 in regulation of hepatic gluconeogenic genes and carbohydrate metabolism in vivo. Nat Med 2004; 10: 168-174.
    • (2004) Nat Med , vol.10 , pp. 168-174
    • Inoue, H.1    Ogawa, W.2    Ozaki, M.3
  • 398
    • 64049109876 scopus 로고    scopus 로고
    • STAT3 inhibition of gluconeogenesis is downregulated by SirT1
    • Nie Y, Erion DM, Yuan Z, et al. STAT3 inhibition of gluconeogenesis is downregulated by SirT1. Nat Cell Biol 2009; 11: 492-500.
    • (2009) Nat Cell Biol , vol.11 , pp. 492-500
    • Nie, Y.1    Erion, D.M.2    Yuan, Z.3
  • 399
    • 0037310863 scopus 로고    scopus 로고
    • RUNX transcription factors as key targets of TGF-beta superfamily signaling
    • Ito Y, Miyazono K. RUNX transcription factors as key targets of TGF-beta superfamily signaling. Curr Opin Genet Dev 2003; 13: 43-47.
    • (2003) Curr Opin Genet Dev , vol.13 , pp. 43-47
    • Ito, Y.1    Miyazono, K.2
  • 400
    • 18344389720 scopus 로고    scopus 로고
    • Causal relationship between the loss of RUNX3 expression and gastric cancer
    • Li QL, Ito K, Sakakura C, et al. Causal relationship between the loss of RUNX3 expression and gastric cancer. Cell 2002; 109: 113-124.
    • (2002) Cell , vol.109 , pp. 113-124
    • Li, Q.L.1    Ito, K.2    Sakakura, C.3
  • 401
    • 33646189483 scopus 로고    scopus 로고
    • RUNX3 cooperates with FoxO3a to induce apoptosis in gastric cancer cells
    • Yamamura Y, Lee WL, Inoue K, Ida H, Ito Y. RUNX3 cooperates with FoxO3a to induce apoptosis in gastric cancer cells. J Biol Chem 2006; 281: 5267-5276.
    • (2006) J Biol Chem , vol.281 , pp. 5267-5276
    • Yamamura, Y.1    Lee, W.L.2    Inoue, K.3    Ida, H.4    Ito, Y.5
  • 402
    • 0038043227 scopus 로고    scopus 로고
    • Smad3 potentiates transforming growth factor beta (TGFbeta)-induced apoptosis and expression of the BH3-only protein Bim in WEHI 231 B lymphocytes
    • Wildey GM, Patil S, Howe PH. Smad3 potentiates transforming growth factor beta (TGFbeta)-induced apoptosis and expression of the BH3-only protein Bim in WEHI 231 B lymphocytes. J Biol Chem 2003; 278: 18069-18077.
    • (2003) J Biol Chem , vol.278 , pp. 18069-18077
    • Wildey, G.M.1    Patil, S.2    Howe, P.H.3
  • 403
    • 67749127709 scopus 로고    scopus 로고
    • Runx1 is a co-activator with FOXO3 to mediate transforming growth factor beta (TGFbeta)-induced Bim transcription in hepatic cells
    • Wildey GM, Howe PH. Runx1 is a co-activator with FOXO3 to mediate transforming growth factor beta (TGFbeta)-induced Bim transcription in hepatic cells. J Biol Chem 2009; 284: 20227-20239.
    • (2009) J Biol Chem , vol.284 , pp. 20227-20239
    • Wildey, G.M.1    Howe, P.H.2
  • 404
    • 0037204948 scopus 로고    scopus 로고
    • TNF-R1 signaling: A beautiful pathway
    • Chen G, Goeddel DV. TNF-R1 signaling: a beautiful pathway. Science 2002; 296: 1634-1635.
    • (2002) Science , vol.296 , pp. 1634-1635
    • Chen, G.1    Goeddel, D.V.2
  • 407
    • 37549050887 scopus 로고    scopus 로고
    • FOXO3a turns the tumor necrosis factor receptor signaling towards apoptosis through reciprocal regulation of c-Jun N-terminal kinase and NF-kappaB
    • Lee HY, Youn SW, Kim JY, et al. FOXO3a turns the tumor necrosis factor receptor signaling towards apoptosis through reciprocal regulation of c-Jun N-terminal kinase and NF-kappaB. Arterioscler Thromb Vasc Biol 2008; 28: 112-120.
    • (2008) Arterioscler Thromb Vasc Biol , vol.28 , pp. 112-120
    • Lee, H.Y.1    Youn, S.W.2    Kim, J.Y.3
  • 408
    • 70349507649 scopus 로고    scopus 로고
    • FoxO4 inhibits NF-kappaB and protects mice against colonic injury and inflammation
    • Zhou W, Cao Q, Peng Y, et al. FoxO4 inhibits NF-kappaB and protects mice against colonic injury and inflammation. Gastroenterology 2009; 137:1403-1414.
    • (2009) Gastroenterology , vol.137 , pp. 1403-1414
    • Zhou, W.1    Cao, Q.2    Peng, Y.3
  • 409
    • 0031005095 scopus 로고    scopus 로고
    • Subtractive cloning and characterization of DRAL, a novel LIM-domain protein down-regulated in rhabdomyosarcoma
    • Genini M, Schwalbe P, Scholl FA, Remppis A, Mattei MG, Schafer BW. Subtractive cloning and characterization of DRAL, a novel LIM-domain protein down-regulated in rhabdomyosarcoma. DNA Cell Biol 1997; 16: 433-442.
    • (1997) DNA Cell Biol , vol.16 , pp. 433-442
    • Genini, M.1    Schwalbe, P.2    Scholl, F.A.3    Remppis, A.4    Mattei, M.G.5    Schafer, B.W.6
  • 410
    • 1642458404 scopus 로고    scopus 로고
    • Extracellular signal-regulated kinase 2 interacts with and is negatively regulated by the LIM-only protein FHL2 in cardiomyocytes
    • Purcell NH, Darwis D, Bueno OF, Muller JM, Schule R, Molkentin JD. Extracellular signal-regulated kinase 2 interacts with and is negatively regulated by the LIM-only protein FHL2 in cardiomyocytes. Mol Cell Biol 2004; 24: 1081-1095.
    • (2004) Mol Cell Biol , vol.24 , pp. 1081-1095
    • Purcell, N.H.1    Darwis, D.2    Bueno, O.F.3    Muller, J.M.4    Schule, R.5    Molkentin, J.D.6
  • 411
    • 0035903468 scopus 로고    scopus 로고
    • Independent function of two destruction domains in hypoxia-inducible factor-alpha chains activated by prolyl hydroxylation
    • Masson N, Willam C, Maxwell PH, Pugh CW, Ratcliffe PJ. Independent function of two destruction domains in hypoxia-inducible factor-alpha chains activated by prolyl hydroxylation. Embo J 2001; 20: 5197-5206.
    • (2001) Embo J , vol.20 , pp. 5197-5206
    • Masson, N.1    Willam, C.2    Maxwell, P.H.3    Pugh, C.W.4    Ratcliffe, P.J.5
  • 412
    • 0035917808 scopus 로고    scopus 로고
    • Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation
    • Jaakkola P, Mole DR, Tian YM, et al. Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 2001; 292: 468-472.
    • (2001) Science , vol.292 , pp. 468-472
    • Jaakkola, P.1    Mole, D.R.2    Tian, Y.M.3
  • 413
    • 0035917313 scopus 로고    scopus 로고
    • HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: Implications for O2 sensing
    • Ivan M, Kondo K, Yang H, et al. HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing. Science 2001; 292: 464-468.
    • (2001) Science , vol.292 , pp. 464-468
    • Ivan, M.1    Kondo, K.2    Yang, H.3
  • 414
    • 40649089427 scopus 로고    scopus 로고
    • PTEN regulates p300-dependent hypoxia-inducible factor 1 transcriptional activity through Forkhead transcription factor 3a (FOXO3a)
    • Emerling BM, Weinberg F, Liu JL, Mak TW, Chandel NS. PTEN regulates p300-dependent hypoxia-inducible factor 1 transcriptional activity through Forkhead transcription factor 3a (FOXO3a). Proc Natl Acad Sci USA 2008; 105: 2622-2627.
    • (2008) Proc Natl Acad Sci USA , vol.105 , pp. 2622-2627
    • Emerling, B.M.1    Weinberg, F.2    Liu, J.L.3    Mak, T.W.4    Chandel, N.S.5
  • 415
    • 0037326985 scopus 로고    scopus 로고
    • PI3K-signalling in B- and T-cells: Insights from gene-targeted mice
    • Okkenhaug K, Vanhaesebroeck B. PI3K-signalling in B- and T-cells: insights from gene-targeted mice. Biochem Soc Trans 2003; 31: 270-274.
    • (2003) Biochem Soc Trans , vol.31 , pp. 270-274
    • Okkenhaug, K.1    Vanhaesebroeck, B.2
  • 416
    • 1342292522 scopus 로고    scopus 로고
    • Phosphoinositide 3-kinase: Diverse roles in immune cell activation
    • Deane JA, Fruman DA. Phosphoinositide 3-kinase: diverse roles in immune cell activation. Annu Rev Immunol 2004; 22: 563-598.
    • (2004) Annu Rev Immunol , vol.22 , pp. 563-598
    • Deane, J.A.1    Fruman, D.A.2
  • 417
    • 2342622123 scopus 로고    scopus 로고
    • Optimal B-cell proliferation requires phosphoinositide 3-kinase-dependent inactivation of FOXO transcription factors
    • Yusuf I, Zhu X, Kharas MG, Chen J, Fruman DA. Optimal B-cell proliferation requires phosphoinositide 3-kinase-dependent inactivation of FOXO transcription factors. Blood 2004; 104: 784-787.
    • (2004) Blood , vol.104 , pp. 784-787
    • Yusuf, I.1    Zhu, X.2    Kharas, M.G.3    Chen, J.4    Fruman, D.A.5
  • 418
    • 0037094096 scopus 로고    scopus 로고
    • The forkhead transcription factor FoxO regulates transcription of p27Kip1 and Bim in response to IL-2
    • Stahl M, Dijkers PF, Kops GJ, et al. The forkhead transcription factor FoxO regulates transcription of p27Kip1 and Bim in response to IL-2. J Immunol 2002; 168: 5024-5031.
    • (2002) J Immunol , vol.168 , pp. 5024-5031
    • Stahl, M.1    Dijkers, P.F.2    Kops, G.J.3
  • 419
    • 33644538251 scopus 로고    scopus 로고
    • FOXO transcription factors cooperate with delta EF1 to activate growth suppressive genes in B lymphocytes
    • Chen J, Yusuf I, Andersen HM, Fruman DA. FOXO transcription factors cooperate with delta EF1 to activate growth suppressive genes in B lymphocytes. J Immunol 2006; 176: 2711-2721.
    • (2006) J Immunol , vol.176 , pp. 2711-2721
    • Chen, J.1    Yusuf, I.2    Andersen, H.M.3    Fruman, D.A.4
  • 421
    • 0036120725 scopus 로고    scopus 로고
    • Control of cell cycle exit and entry by protein kinase B-regulated forkhead transcription factors
    • Kops GJ, Medema RH, Glassford J, et al. Control of cell cycle exit and entry by protein kinase B-regulated forkhead transcription factors. Mol Cell Biol 2002; 22: 2025-2036.
    • (2002) Mol Cell Biol , vol.22 , pp. 2025-2036
    • Kops, G.J.1    Medema, R.H.2    Glassford, J.3
  • 422
    • 0035193044 scopus 로고    scopus 로고
    • The transcriptional repressor ZEB regulates p73 expression at the crossroad between proliferation and differentiation
    • Fontemaggi G, Gurtner A, Strano S, et al. The transcriptional repressor ZEB regulates p73 expression at the crossroad between proliferation and differentiation. Mol Cell Biol 2001; 21: 8461-8470.
    • (2001) Mol Cell Biol , vol.21 , pp. 8461-8470
    • Fontemaggi, G.1    Gurtner, A.2    Strano, S.3
  • 423
    • 0038324070 scopus 로고    scopus 로고
    • Regulation of Smad signaling through a differential recruitment of coactivators and corepressors by ZEB proteins
    • Postigo AA, Depp JL, Taylor JJ, Kroll KL. Regulation of Smad signaling through a differential recruitment of coactivators and corepressors by ZEB proteins. Embo J 2003; 22: 2453-2462.
    • (2003) Embo J , vol.22 , pp. 2453-2462
    • Postigo, A.A.1    Depp, J.L.2    Taylor, J.J.3    Kroll, K.L.4
  • 424
    • 0032080450 scopus 로고    scopus 로고
    • Transcriptional repression of the IL-2 gene in Th cells by ZEB
    • Yasui DH, Genetta T, Kadesch T, et al. Transcriptional repression of the IL-2 gene in Th cells by ZEB. J Immunol 1998; 160: 4433-4440.
    • (1998) J Immunol , vol.160 , pp. 4433-4440
    • Yasui, D.H.1    Genetta, T.2    Kadesch, T.3
  • 425
    • 0030906128 scopus 로고    scopus 로고
    • Impairment of T cell development in deltaEF1 mutant mice
    • Higashi Y, Moribe H, Takagi T, et al. Impairment of T cell development in deltaEF1 mutant mice. J Exp Med 1997; 185: 1467-1479.
    • (1997) J Exp Med , vol.185 , pp. 1467-1479
    • Higashi, Y.1    Moribe, H.2    Takagi, T.3
  • 426
    • 0034716888 scopus 로고    scopus 로고
    • A genetic link between morphogenesis and cell division during formation of the ventral furrow in Drosophila
    • Grosshans J, Wieschaus E. A genetic link between morphogenesis and cell division during formation of the ventral furrow in Drosophila. Cell 2000; 101: 523-531.
    • (2000) Cell , vol.101 , pp. 523-531
    • Grosshans, J.1    Wieschaus, E.2
  • 427
    • 77952672913 scopus 로고    scopus 로고
    • Human TRIB2 is a repressor of FOXO that contributes to the malignant phenotype of melanoma cells
    • Zanella F, Renner O, Garcia B, et al. Human TRIB2 is a repressor of FOXO that contributes to the malignant phenotype of melanoma cells. Oncogene 2010; 29: 2973-2982.
    • (2010) Oncogene , vol.29 , pp. 2973-2982
    • Zanella, F.1    Renner, O.2    Garcia, B.3
  • 428
    • 0034463096 scopus 로고    scopus 로고
    • Follicle-Stimulating hormone (FSH) stimulates phosphorylation and activation of protein kinase B (PKB/Akt) and serum and glucocorticoid-lnduced kinase (Sgk): Evidence for A kinase-independent signaling by FSH in granulosa cells
    • Gonzalez-Robayna IJ, Falender AE, Ochsner S, Firestone GL, Richards JS. Follicle-Stimulating hormone (FSH) stimulates phosphorylation and activation of protein kinase B (PKB/Akt) and serum and glucocorticoid-lnduced kinase (Sgk): evidence for A kinase-independent signaling by FSH in granulosa cells. Mol Endocrinol 2000; 14: 1283-1300.
    • (2000) Mol Endocrinol , vol.14 , pp. 1283-1300
    • Gonzalez-Robayna, I.J.1    Falender, A.E.2    Ochsner, S.3    Firestone, G.L.4    Richards, J.S.5
  • 429
    • 2942601056 scopus 로고    scopus 로고
    • Human follicle-stimulating hormone (FSH) receptor interacts with the adaptor protein APPL1 in HEK 293 cells: Potential involvement of the PI3K pathway in FSH signaling
    • Nechamen CA, Thomas RM, Cohen BD, et al. Human follicle-stimulating hormone (FSH) receptor interacts with the adaptor protein APPL1 in HEK 293 cells: potential involvement of the PI3K pathway in FSH signaling. Biol Reprod 2004; 71:629-636.
    • (2004) Biol Reprod , vol.71 , pp. 629-636
    • Nechamen, C.A.1    Thomas, R.M.2    Cohen, B.D.3
  • 430
    • 33751239461 scopus 로고    scopus 로고
    • APPL1, APPL2, Akt2 and FOXO1a interact with FSHR in a potential signaling complex
    • Nechamen CA, Thomas RM, Dias JA. APPL1, APPL2, Akt2 and FOXO1a interact with FSHR in a potential signaling complex. Mol Cell Endocrinol 2007; 260-262: 93-99.
    • (2007) Mol Cell Endocrinol , vol.260-262 , pp. 93-99
    • Nechamen, C.A.1    Thomas, R.M.2    Dias, J.A.3
  • 431
    • 2442485795 scopus 로고    scopus 로고
    • Follicle-stimulating hormone activation of hypoxia-inducible factor-1 by the phosphatidylinositol 3-kinase/AKT/Ras homolog enriched in brain (Rheb)/mammalian target of rapamycin (mTOR) pathway is necessary for induction of select protein markers of follicular differentiation
    • Alam H, Maizels ET, Park Y, et al. Follicle-stimulating hormone activation of hypoxia-inducible factor-1 by the phosphatidylinositol 3-kinase/AKT/Ras homolog enriched in brain (Rheb)/mammalian target of rapamycin (mTOR) pathway is necessary for induction of select protein markers of follicular differentiation. J Biol Chem 2004; 279: 19431-19440.
    • (2004) J Biol Chem , vol.279 , pp. 19431-19440
    • Alam, H.1    Maizels, E.T.2    Park, Y.3
  • 432
    • 18344387559 scopus 로고    scopus 로고
    • Phosphorylation-specific prolyl isomerization: Is there an underlying theme?
    • Wulf G, Finn G, Suizu F, Lu KP. Phosphorylation-specific prolyl isomerization: is there an underlying theme? Nat Cell Biol 2005; 7: 435-441.
    • (2005) Nat Cell Biol , vol.7 , pp. 435-441
    • Wulf, G.1    Finn, G.2    Suizu, F.3    Lu, K.P.4
  • 433
    • 0037351048 scopus 로고    scopus 로고
    • Prolyl isomerase Pin1: A catalyst for oncogenesis and a potential therapeutic target in cancer
    • Ryo A, Liou YC, Lu KP, Wulf G. Prolyl isomerase Pin1: a catalyst for oncogenesis and a potential therapeutic target in cancer. J Cell Sci 2003; 116: 773-783.
    • (2003) J Cell Sci , vol.116 , pp. 773-783
    • Ryo, A.1    Liou, Y.C.2    Lu, K.P.3    Wulf, G.4
  • 434
    • 54749134224 scopus 로고    scopus 로고
    • The peptidyl-isomerase Pin1 regulates p27kip1 expression through inhibition of Forkhead box O tumor suppressors
    • Brenkman AB, de Keizer PL, van den Broek NJ, et al. The peptidyl-isomerase Pin1 regulates p27kip1 expression through inhibition of Forkhead box O tumor suppressors. Cancer Res 2008; 68: 7597-605.
    • (2008) Cancer Res , vol.68 , pp. 7597-7605
    • Brenkman, A.B.1    de Keizer, P.L.2    van den Broek, N.J.3
  • 436
    • 0028922462 scopus 로고
    • Mice lacking ADPRT and poly(ADP-ribosyl)ation develop normally but are susceptible to skin disease
    • Wang ZQ, Auer B, Stingl L, et al. Mice lacking ADPRT and poly(ADP-ribosyl)ation develop normally but are susceptible to skin disease. Genes Dev 1995; 9: 509-520.
    • (1995) Genes Dev , vol.9 , pp. 509-520
    • Wang, Z.Q.1    Auer, B.2    Stingl, L.3
  • 437
    • 34548183836 scopus 로고    scopus 로고
    • PolyADP-ribosylation and cancer
    • Miwa M, Masutani M. PolyADP-ribosylation and cancer. Cancer Sci 2007; 98: 1528-1535.
    • (2007) Cancer Sci , vol.98 , pp. 1528-1535
    • Miwa, M.1    Masutani, M.2
  • 438
    • 10944253639 scopus 로고    scopus 로고
    • Activating the PARP-1 sensor component of the groucho/ TLE1 corepressor complex mediates a CaMKinase IIdelta-dependent neurogenic gene activation pathway
    • Ju BG, Solum D, Song EJ, et al. Activating the PARP-1 sensor component of the groucho/ TLE1 corepressor complex mediates a CaMKinase IIdelta-dependent neurogenic gene activation pathway. Cell 2004; 119: 815-829.
    • (2004) Cell , vol.119 , pp. 815-829
    • Ju, B.G.1    Solum, D.2    Song, E.J.3
  • 439
    • 44649130038 scopus 로고    scopus 로고
    • Transcriptional control by PARP-1: Chromatin modulation, enhancer-binding, coregulation, and insulation
    • Kraus WL. Transcriptional control by PARP-1: chromatin modulation, enhancer-binding, coregulation, and insulation. Curr Opin Cell Biol 2008; 20: 294-302.
    • (2008) Curr Opin Cell Biol , vol.20 , pp. 294-302
    • Kraus, W.L.1


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.