메뉴 건너뛰기




Volumn 95, Issue 4, 2015, Pages 324-336

Mechanisms of tolvaptan-induced toxicity in HepG2 cells

Author keywords

Apoptosis; Autophagy; DNA damage; Liver injury; Proteasome; Tolvaptan

Indexed keywords

8 HYDROXYGUANINE; CYCLIN DEPENDENT KINASE; CYCLINE; CYTOCHROME C; HISTONE H2AX; PROTEIN BCL 2; SYNAPTOPHYSIN; TOLVAPTAN; BENZAZEPINE DERIVATIVE; CYCLIN D3; CYCLIN DEPENDENT KINASE 4; CYTOCHROME P450 3A; MITOGEN ACTIVATED PROTEIN KINASE; PROTEASOME; PROTEASOME INHIBITOR; VASOPRESSIN RECEPTOR ANTAGONIST;

EID: 84929705874     PISSN: 00062952     EISSN: 18732968     Source Type: Journal    
DOI: 10.1016/j.bcp.2015.03.015     Document Type: Article
Times cited : (29)

References (65)
  • 1
    • 84875397423 scopus 로고    scopus 로고
    • 2-receptor antagonist tolvaptan
    • 2-receptor antagonist tolvaptan J. Crit. Care 28 219 2013 e1 e12
    • (2013) J. Crit. Care , vol.28 , Issue.219 , pp. e1-e12
    • Friedman, B.1    Cirulli, J.2
  • 4
    • 84857048525 scopus 로고    scopus 로고
    • Effect of grapefruit juice on the pharmacokinetics of tolvaptan, a non-peptide arginine vasopressin antagonist, in healthy subjects
    • S.E. Shoaf, S. Mallikaarjun, and P. Bricmont Effect of grapefruit juice on the pharmacokinetics of tolvaptan, a non-peptide arginine vasopressin antagonist, in healthy subjects Eur. J. Clin. Pharmacol. 68 2012 207 211
    • (2012) Eur. J. Clin. Pharmacol. , vol.68 , pp. 207-211
    • Shoaf, S.E.1    Mallikaarjun, S.2    Bricmont, P.3
  • 5
    • 84858202663 scopus 로고    scopus 로고
    • Effects of CYP3A4 inhibition and induction on the pharmacokinetics and pharmacodynamics of tolvaptan, a non-peptide AVP antagonist in healthy subjects
    • S.E. Shoaf, P. Bricmont, and S. Mallikaarjun Effects of CYP3A4 inhibition and induction on the pharmacokinetics and pharmacodynamics of tolvaptan, a non-peptide AVP antagonist in healthy subjects Br. J. Clin. Pharmacol. 73 2012 579 587
    • (2012) Br. J. Clin. Pharmacol. , vol.73 , pp. 579-587
    • Shoaf, S.E.1    Bricmont, P.2    Mallikaarjun, S.3
  • 6
    • 77949464718 scopus 로고    scopus 로고
    • From basic research to clinical development of MEK1/2 inhibitors for cancer therapy
    • C. Frémin, and S. Meloche From basic research to clinical development of MEK1/2 inhibitors for cancer therapy J. Hematol. Oncol. 3 2010 8
    • (2010) J. Hematol. Oncol. , vol.3 , pp. 8
    • Frémin, C.1    Meloche, S.2
  • 7
    • 84855795110 scopus 로고    scopus 로고
    • MEK1/2 dual-specificity protein kinases: Structure and regulation
    • R. Roskoski Jr. MEK1/2 dual-specificity protein kinases: structure and regulation Biochem. Biophys. Res. Commun. 417 2012 5 10
    • (2012) Biochem. Biophys. Res. Commun. , vol.417 , pp. 5-10
    • Roskoski, Jr.R.1
  • 8
    • 0030924833 scopus 로고    scopus 로고
    • Pyridinyl imidazole inhibitors of p38 mitogen-activated protein kinase bind in the ATP site
    • P.R. Young, M.M. McLaughlin, S. Kumar, S. Kassis, M.L. Doyle, and D. McNulty Pyridinyl imidazole inhibitors of p38 mitogen-activated protein kinase bind in the ATP site J. Biol. Chem. 272 1997 12116 12121
    • (1997) J. Biol. Chem. , vol.272 , pp. 12116-12121
    • Young, P.R.1    McLaughlin, M.M.2    Kumar, S.3    Kassis, S.4    Doyle, M.L.5    McNulty, D.6
  • 9
    • 0029877917 scopus 로고    scopus 로고
    • Differential inhibition of calpain and proteasome activities by peptidyl aldehydes of di-leucine and tri-leucine
    • S. Tsubuki, Y. Saito, M. Tomioka, H. Ito, and S. Kawashima Differential inhibition of calpain and proteasome activities by peptidyl aldehydes of di-leucine and tri-leucine J. Biochem. 119 1996 572 576
    • (1996) J. Biochem. , vol.119 , pp. 572-576
    • Tsubuki, S.1    Saito, Y.2    Tomioka, M.3    Ito, H.4    Kawashima, S.5
  • 10
    • 84901922071 scopus 로고    scopus 로고
    • Sertraline induces endoplasmic reticulum stress in hepatic cells
    • S. Chen, J. Xuan, L. Couch, A. Iyer, Y. Wu, and Q.-Z. Li Sertraline induces endoplasmic reticulum stress in hepatic cells Toxicology 322 2014 78 88
    • (2014) Toxicology , vol.322 , pp. 78-88
    • Chen, S.1    Xuan, J.2    Couch, L.3    Iyer, A.4    Wu, Y.5    Li, Q.-Z.6
  • 11
    • 79951922704 scopus 로고    scopus 로고
    • Similarities and differences in the expression of drug-metabolizing enzymes between human hepatic cell lines and primary human hepatocytes
    • L. Guo, S. Dial, L. Shi, W. Branham, J. Liu, and J.-L. Fang Similarities and differences in the expression of drug-metabolizing enzymes between human hepatic cell lines and primary human hepatocytes Drug Metab. Dispos. 39 2011 528 538
    • (2011) Drug Metab. Dispos. , vol.39 , pp. 528-538
    • Guo, L.1    Dial, S.2    Shi, L.3    Branham, W.4    Liu, J.5    Fang, J.-L.6
  • 12
    • 84927762227 scopus 로고    scopus 로고
    • The role of autophagy in usnic acidinduced toxicity in hepatic cells
    • S. Chen, V.N. Dobrovolsky, F. Liu, Y. Wu, Z. Zhang, and N. Mei The role of autophagy in usnic acidinduced toxicity in hepatic cells Toxicol. Sci. 142 2014 33 44
    • (2014) Toxicol. Sci. , vol.142 , pp. 33-44
    • Chen, S.1    Dobrovolsky, V.N.2    Liu, F.3    Wu, Y.4    Zhang, Z.5    Mei, N.6
  • 13
    • 84937970242 scopus 로고    scopus 로고
    • Extracellular signal-regulated kinases 1/2 and Akt contribute to triclosan-stimulated proliferation of JB6 Cl 41-5a cells
    • Y. Wu, F.A. Beland, S. Chen, and J.-L. Fang Extracellular signal-regulated kinases 1/2 and Akt contribute to triclosan-stimulated proliferation of JB6 Cl 41-5a cells Arch. Toxicol. 2014 10.1007/s00204-014-1308-5
    • (2014) Arch. Toxicol.
    • Wu, Y.1    Beland, F.A.2    Chen, S.3    Fang, J.-L.4
  • 14
    • 69049092784 scopus 로고    scopus 로고
    • Long-term exposure to zidovudine delays cell cycle progression, induces apoptosis, and decreases telomerase activity in human hepatocytes
    • J.-L. Fang, and F.A. Beland Long-term exposure to zidovudine delays cell cycle progression, induces apoptosis, and decreases telomerase activity in human hepatocytes Toxicol. Sci. 111 2009 120 130
    • (2009) Toxicol. Sci. , vol.111 , pp. 120-130
    • Fang, J.-L.1    Beland, F.A.2
  • 15
    • 0036142218 scopus 로고    scopus 로고
    • Mitotic phosphorylation of histone H3: Spatiooral regulation by mammalian Aurora kinases
    • C. Crosio, G.M. Fimia, R. Loury, M. Kimura, Y. Okano, and H. Zhou Mitotic phosphorylation of histone H3: spatiooral regulation by mammalian Aurora kinases Mol. Cell Biol. 22 2002 874 885
    • (2002) Mol. Cell Biol. , vol.22 , pp. 874-885
    • Crosio, C.1    Fimia, G.M.2    Loury, R.3    Kimura, M.4    Okano, Y.5    Zhou, H.6
  • 16
    • 84859731654 scopus 로고    scopus 로고
    • Identification of BC005512 as a DNA damage responsive murine endogenous retrovirus of GLN family involved in cell growth regulation
    • Y. Wu, X. Qi, L. Gong, G. Xing, M. Chen, and L. Miao Identification of BC005512 as a DNA damage responsive murine endogenous retrovirus of GLN family involved in cell growth regulation PLoS ONE 7 2012 e35010
    • (2012) PLoS ONE , vol.7 , pp. e35010
    • Wu, Y.1    Qi, X.2    Gong, L.3    Xing, G.4    Chen, M.5    Miao, L.6
  • 17
    • 84908287538 scopus 로고    scopus 로고
    • Differential effects of triclosan on the activation of mouse and human peroxisome proliferator-activated receptor alpha
    • Y. Wu, Q. Wu, F.A. Beland, P. Ge, M.G. Manjanatha, and J.-L. Fang Differential effects of triclosan on the activation of mouse and human peroxisome proliferator-activated receptor alpha Toxicol. Lett. 231 2014 17 28
    • (2014) Toxicol. Lett. , vol.231 , pp. 17-28
    • Wu, Y.1    Wu, Q.2    Beland, F.A.3    Ge, P.4    Manjanatha, M.G.5    Fang, J.-L.6
  • 18
    • 0038052805 scopus 로고    scopus 로고
    • The cell cycle: A review of regulation, deregulation and therapeutic targets in cancer
    • K. Vermeulen, D.R. Van Bockstaele, and Z.N. Berneman The cell cycle: a review of regulation, deregulation and therapeutic targets in cancer Cell Prolif. 36 2003 131 149
    • (2003) Cell Prolif. , vol.36 , pp. 131-149
    • Vermeulen, K.1    Van Bockstaele, D.R.2    Berneman, Z.N.3
  • 19
    • 0033711708 scopus 로고    scopus 로고
    • 1 cyclin-dependent kinases in the mammalian cell cycle
    • 1 cyclin-dependent kinases in the mammalian cell cycle Curr. Opin. Cell Biol. 12 2000 676 684
    • (2000) Curr. Opin. Cell Biol. , vol.12 , pp. 676-684
    • Ekholm, S.V.1    Reed, S.I.2
  • 21
    • 0032489520 scopus 로고    scopus 로고
    • DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139
    • E.P. Rogakou, D.R. Pilch, A.H. Orr, V.S. Ivanova, and W.M. Bonner DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139 J. Biol. Chem. 273 1998 5858 5868
    • (1998) J. Biol. Chem. , vol.273 , pp. 5858-5868
    • Rogakou, E.P.1    Pilch, D.R.2    Orr, A.H.3    Ivanova, V.S.4    Bonner, W.M.5
  • 22
    • 0035449355 scopus 로고    scopus 로고
    • Cell cycle checkpoint signaling through the ATM and ATR kinases
    • R.T. Abraham Cell cycle checkpoint signaling through the ATM and ATR kinases Genes Dev. 15 2001 2177 2196
    • (2001) Genes Dev. , vol.15 , pp. 2177-2196
    • Abraham, R.T.1
  • 23
    • 0035831473 scopus 로고    scopus 로고
    • Executioner caspase-3, -6, and -7 perform distinct, non-redundant roles during the demolition phase of apoptosis
    • E.A. Slee, C. Adrain, and S.J. Martin Executioner caspase-3, -6, and -7 perform distinct, non-redundant roles during the demolition phase of apoptosis J. Biol. Chem. 276 2001 7320 7326
    • (2001) J. Biol. Chem. , vol.276 , pp. 7320-7326
    • Slee, E.A.1    Adrain, C.2    Martin, S.J.3
  • 24
    • 0842281645 scopus 로고    scopus 로고
    • Cell death: Critical control points
    • N.N. Danial, and S.J. Korsmeyer Cell death: critical control points Cell 116 2004 205 219
    • (2004) Cell , vol.116 , pp. 205-219
    • Danial, N.N.1    Korsmeyer, S.J.2
  • 25
    • 77956095537 scopus 로고    scopus 로고
    • Mitochondria and cell death: Outer membrane permeabilization and beyond
    • S.W.G. Tait, and D.R. Green Mitochondria and cell death: outer membrane permeabilization and beyond Nat. Rev. Mol. Cell Biol. 11 2010 621 632
    • (2010) Nat. Rev. Mol. Cell Biol. , vol.11 , pp. 621-632
    • Tait, S.W.G.1    Green, D.R.2
  • 27
    • 0031918223 scopus 로고    scopus 로고
    • BCL-2 family: Regulators of cell death
    • D.T. Chao, and S.J. Korsmeyer BCL-2 family: regulators of cell death Annu. Rev. Immunol. 16 1998 395 419
    • (1998) Annu. Rev. Immunol. , vol.16 , pp. 395-419
    • Chao, D.T.1    Korsmeyer, S.J.2
  • 28
    • 0033179760 scopus 로고    scopus 로고
    • BCL-2 family members and the mitochondria in apoptosis
    • A. Gross, J.M. McDonnell, and S.J. Korsmeyer BCL-2 family members and the mitochondria in apoptosis Genes Dev. 13 1999 1899 1911
    • (1999) Genes Dev. , vol.13 , pp. 1899-1911
    • Gross, A.1    McDonnell, J.M.2    Korsmeyer, S.J.3
  • 29
    • 79960230433 scopus 로고    scopus 로고
    • Mitochondria in apoptosis: Bcl-2 family members and mitochondrial dynamics
    • J.-C. Martinou, and R.J. Youle Mitochondria in apoptosis: Bcl-2 family members and mitochondrial dynamics Dev. Cell. 21 2011 92 101
    • (2011) Dev. Cell. , vol.21 , pp. 92-101
    • Martinou, J.-C.1    Youle, R.J.2
  • 30
    • 79952112019 scopus 로고    scopus 로고
    • The MAP kinase signaling cascades: A system of hundreds of components regulates a diverse array of physiological functions
    • (MAP Kinase Signaling Protocols, Methods in Molecular Biology)
    • Y. Keshet, and R. Seger The MAP kinase signaling cascades: a system of hundreds of components regulates a diverse array of physiological functions Methods Mol. Biol. 661 2010 3 38 (MAP Kinase Signaling Protocols, Methods in Molecular Biology)
    • (2010) Methods Mol. Biol. , vol.661 , pp. 3-38
    • Keshet, Y.1    Seger, R.2
  • 32
    • 0035282334 scopus 로고    scopus 로고
    • Mammalian MAP kinase signalling cascades
    • L. Chang, and M. Karin Mammalian MAP kinase signalling cascades Nature 410 2001 37 40
    • (2001) Nature , vol.410 , pp. 37-40
    • Chang, L.1    Karin, M.2
  • 33
    • 33646345376 scopus 로고    scopus 로고
    • Ubiquitin ligases: Cell-cycle control and cancer
    • K.I. Nakayama, and K. Nakayama Ubiquitin ligases: cell-cycle control and cancer Nat. Rev. Cancer 6 2006 369 381
    • (2006) Nat. Rev. Cancer , vol.6 , pp. 369-381
    • Nakayama, K.I.1    Nakayama, K.2
  • 34
    • 84878934964 scopus 로고    scopus 로고
    • Ubiquitin ligases and cell cycle control
    • L.K. Teixeira, and S.I. Reed Ubiquitin ligases and cell cycle control Annu. Rev. Biochem. 82 2013 387 414
    • (2013) Annu. Rev. Biochem. , vol.82 , pp. 387-414
    • Teixeira, L.K.1    Reed, S.I.2
  • 35
    • 0032919345 scopus 로고    scopus 로고
    • The role of the ubiquitin-proteasome pathway in apoptosis
    • R.Z. Orlowski The role of the ubiquitin-proteasome pathway in apoptosis Cell Death Differ. 6 1999 303 313
    • (1999) Cell Death Differ. , vol.6 , pp. 303-313
    • Orlowski, R.Z.1
  • 36
    • 0036355705 scopus 로고    scopus 로고
    • Regulation of apoptosis by the ubiquitin and proteasome pathway
    • C. Wójcik Regulation of apoptosis by the ubiquitin and proteasome pathway J. Cell. Mol. Med. 6 2002 25 48
    • (2002) J. Cell. Mol. Med. , vol.6 , pp. 25-48
    • Wójcik, C.1
  • 37
    • 70449091753 scopus 로고    scopus 로고
    • Stepwise activation of BAX and BAK by tBID, BIM, and PUMA initiates mitochondrial apoptosis
    • H. Kim, H.-C. Tu, D. Ren, O. Takeuchi, J.R. Jeffers, and G.P. Zambetti Stepwise activation of BAX and BAK by tBID, BIM, and PUMA initiates mitochondrial apoptosis Mol. Cell 36 2009 487 499
    • (2009) Mol. Cell , vol.36 , pp. 487-499
    • Kim, H.1    Tu, H.-C.2    Ren, D.3    Takeuchi, O.4    Jeffers, J.R.5    Zambetti, G.P.6
  • 38
    • 0032555697 scopus 로고    scopus 로고
    • Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis
    • H. Li, H. Zhu, C.-J. Xu, and J. Yuan Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis Cell 94 1998 491 501
    • (1998) Cell , vol.94 , pp. 491-501
    • Li, H.1    Zhu, H.2    Xu, C.-J.3    Yuan, J.4
  • 39
    • 0034193138 scopus 로고    scopus 로고
    • Cleavage of Bax enhances its cell death function
    • D.E. Wood, and E.W. Newcomb Cleavage of Bax enhances its cell death function Exp. Cell Res. 256 2000 375 382
    • (2000) Exp. Cell Res. , vol.256 , pp. 375-382
    • Wood, D.E.1    Newcomb, E.W.2
  • 40
    • 37649005234 scopus 로고    scopus 로고
    • Autophagy in the pathogenesis of disease
    • B. Levine, and G. Kroemer Autophagy in the pathogenesis of disease Cell 132 2008 27 42
    • (2008) Cell , vol.132 , pp. 27-42
    • Levine, B.1    Kroemer, G.2
  • 41
    • 75749122303 scopus 로고    scopus 로고
    • Methods in mammalian autophagy research
    • N. Mizushima, T. Yoshimori, and B. Levine Methods in mammalian autophagy research Cell 140 2010 313 326
    • (2010) Cell , vol.140 , pp. 313-326
    • Mizushima, N.1    Yoshimori, T.2    Levine, B.3
  • 42
    • 20344387475 scopus 로고    scopus 로고
    • Autophagy: Dual roles in life and death
    • E.H. Baehrecke Autophagy: dual roles in life and death Nat. Rev. Mol. Cell Biol. 6 2005 505 510
    • (2005) Nat. Rev. Mol. Cell Biol. , vol.6 , pp. 505-510
    • Baehrecke, E.H.1
  • 43
    • 80655124381 scopus 로고    scopus 로고
    • Autophagy as a rescue mechanism in Efavirenz-induced mitochondrial dysfunction: A lesson from hepatic cells
    • N. Apostolova, L.J. Gomez-Sucerquia, A. Gortat, A. Blas-Garcia, and J.V. Esplugues Autophagy as a rescue mechanism in Efavirenz-induced mitochondrial dysfunction: a lesson from hepatic cells Autophagy 7 2011 1402 1404
    • (2011) Autophagy , vol.7 , pp. 1402-1404
    • Apostolova, N.1    Gomez-Sucerquia, L.J.2    Gortat, A.3    Blas-Garcia, A.4    Esplugues, J.V.5
  • 45
    • 84155194923 scopus 로고    scopus 로고
    • Activation of autophagy protects against acetaminophen-induced hepatotoxicity
    • H.-M. Ni, A. Bockus, N. Boggess, H. Jaeschke, and W.-X. Ding Activation of autophagy protects against acetaminophen-induced hepatotoxicity Hepatology 55 2012 222 231
    • (2012) Hepatology , vol.55 , pp. 222-231
    • Ni, H.-M.1    Bockus, A.2    Boggess, N.3    Jaeschke, H.4    Ding, W.-X.5
  • 46
    • 0034329418 scopus 로고    scopus 로고
    • LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing
    • Y. Kabeya, N. Mizushima, T. Ueno, A. Yamamoto, T. Kirisako, and T. Noda LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing EMBO J. 19 2000 5720 5728
    • (2000) EMBO J. , vol.19 , pp. 5720-5728
    • Kabeya, Y.1    Mizushima, N.2    Ueno, T.3    Yamamoto, A.4    Kirisako, T.5    Noda, T.6
  • 47
    • 46849121421 scopus 로고    scopus 로고
    • Utilizing flow cytometry to monitor autophagy in living mammalian cells
    • E. Shvets, E. Fass, and Z. Elazar Utilizing flow cytometry to monitor autophagy in living mammalian cells Autophagy 4 2008 621 628
    • (2008) Autophagy , vol.4 , pp. 621-628
    • Shvets, E.1    Fass, E.2    Elazar, Z.3
  • 48
    • 21044455137 scopus 로고    scopus 로고
    • Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice
    • M. Komatsu, S. Waguri, T. Ueno, J. Iwata, S. Murata, and I. Tanida Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice J. Cell Biol. 169 2005 425 434
    • (2005) J. Cell Biol. , vol.169 , pp. 425-434
    • Komatsu, M.1    Waguri, S.2    Ueno, T.3    Iwata, J.4    Murata, S.5    Tanida, I.6
  • 49
    • 43349090542 scopus 로고    scopus 로고
    • In vitro assessment of mitochondrial dysfunction and cytotoxicity of nefazodone, trazodone, and buspirone
    • J.A. Dykens, J.D. Jamieson, L.D. Marroquin, S. Nadanaciva, J.J. Xu, and M.C. Dunn In vitro assessment of mitochondrial dysfunction and cytotoxicity of nefazodone, trazodone, and buspirone Toxicol. Sci. 103 2008 335 345
    • (2008) Toxicol. Sci. , vol.103 , pp. 335-345
    • Dykens, J.A.1    Jamieson, J.D.2    Marroquin, L.D.3    Nadanaciva, S.4    Xu, J.J.5    Dunn, M.C.6
  • 50
    • 84873821138 scopus 로고    scopus 로고
    • Mechanisms of hepatocellular toxicity associated with dronedarone - A comparison to amiodarone
    • A. Felser, K. Blum, P.W. Lindinger, J. Bouitbir, and S. Krähenbühl Mechanisms of hepatocellular toxicity associated with dronedarone - a comparison to amiodarone Toxicol. Sci. 131 2013 480 490
    • (2013) Toxicol. Sci. , vol.131 , pp. 480-490
    • Felser, A.1    Blum, K.2    Lindinger, P.W.3    Bouitbir, J.4    Krähenbühl, S.5
  • 51
    • 77049121038 scopus 로고    scopus 로고
    • Cell based approaches for evaluation of drug-induced liver injury
    • M.L. Greer, J. Barber, J. Eakins, and J.G. Kenna Cell based approaches for evaluation of drug-induced liver injury Toxicology 268 2010 125 131
    • (2010) Toxicology , vol.268 , pp. 125-131
    • Greer, M.L.1    Barber, J.2    Eakins, J.3    Kenna, J.G.4
  • 52
    • 84874684601 scopus 로고    scopus 로고
    • Involvement of enniatins-induced cytotoxicity in human HepG2 cells
    • A. Juan-García, L. Manyes, M.-J. Ruiz, and G. Font Involvement of enniatins-induced cytotoxicity in human HepG2 cells Toxicol. Lett. 218 2013 166 173
    • (2013) Toxicol. Lett. , vol.218 , pp. 166-173
    • Juan-García, A.1    Manyes, L.2    Ruiz, M.-J.3    Font, G.4
  • 53
    • 84875249687 scopus 로고    scopus 로고
    • Cadmium telluride quantum dots cause oxidative stress leading to extrinsic and intrinsic apoptosis in hepatocellular carcinoma HepG2 cells
    • K.C. Nguyen, W.G. Willmore, and A.F. Tayabali Cadmium telluride quantum dots cause oxidative stress leading to extrinsic and intrinsic apoptosis in hepatocellular carcinoma HepG2 cells Toxicology 306 2013 114 123
    • (2013) Toxicology , vol.306 , pp. 114-123
    • Nguyen, K.C.1    Willmore, W.G.2    Tayabali, A.F.3
  • 54
    • 45549089681 scopus 로고    scopus 로고
    • Estrogen and progesterone lower cyclin B1 and D1 expression, block cell cycle in G2/M, and trigger apoptosis in human adrenal carcinoma cell cultures
    • J.W. Brown, L.M. Prieto, C. Perez-Stable, M. Montoya, S. Cappell, and L.M. Fishman Estrogen and progesterone lower cyclin B1 and D1 expression, block cell cycle in G2/M, and trigger apoptosis in human adrenal carcinoma cell cultures Horm. Metab. Res. 40 2008 306 310
    • (2008) Horm. Metab. Res. , vol.40 , pp. 306-310
    • Brown, J.W.1    Prieto, L.M.2    Perez-Stable, C.3    Montoya, M.4    Cappell, S.5    Fishman, L.M.6
  • 55
    • 79952327221 scopus 로고    scopus 로고
    • Cantharidin induces G2/M phase arrest and apoptosis in human colorectal cancer colo 205 cells through inhibition of CDK1 activity and caspase-dependent signaling pathways
    • W.-W. Huang, S.-W. Ko, H.-Y. Tsai, J.-G. Chung, J.-H. Chiang, and K.-T. Chen Cantharidin induces G2/M phase arrest and apoptosis in human colorectal cancer colo 205 cells through inhibition of CDK1 activity and caspase-dependent signaling pathways Int. J. Oncol. 38 2011 1067 1073
    • (2011) Int. J. Oncol. , vol.38 , pp. 1067-1073
    • Huang, W.-W.1    Ko, S.-W.2    Tsai, H.-Y.3    Chung, J.-G.4    Chiang, J.-H.5    Chen, K.-T.6
  • 57
    • 84880308946 scopus 로고    scopus 로고
    • 2/M arrest by inhibiting CDK1 activity and causing apoptosis via ROS-mitochondrial apoptotic pathway in human hepatocarcinoma cells in vitro and in vivo
    • 2/M arrest by inhibiting CDK1 activity and causing apoptosis via ROS-mitochondrial apoptotic pathway in human hepatocarcinoma cells in vitro and in vivo Carcinogenesis 34 2013 1636 1643
    • (2013) Carcinogenesis , vol.34 , pp. 1636-1643
    • Wu, W.1    Ye, H.2    Wan, L.3    Han, X.4    Wang, G.5    Hu, J.6
  • 58
    • 79951702955 scopus 로고    scopus 로고
    • Bortezomib as the first proteasome inhibitor anticancer drug: Current status and future perspectives
    • D. Chen, M. Frezza, S. Schmitt, J. Kanwar, and Q.P. Dou Bortezomib as the first proteasome inhibitor anticancer drug: current status and future perspectives Curr. Cancer Drug Targets 11 2011 239 253
    • (2011) Curr. Cancer Drug Targets , vol.11 , pp. 239-253
    • Chen, D.1    Frezza, M.2    Schmitt, S.3    Kanwar, J.4    Dou, Q.P.5
  • 59
    • 79956071998 scopus 로고    scopus 로고
    • Carfilzomib: A novel second-generation proteasome inhibitor
    • M.L. Khan, and A.K. Stewart Carfilzomib: a novel second-generation proteasome inhibitor Future Oncol. 7 2011 607 612
    • (2011) Future Oncol. , vol.7 , pp. 607-612
    • Khan, M.L.1    Stewart, A.K.2
  • 61
    • 84871306932 scopus 로고    scopus 로고
    • Pharmacokinetics and pharmacodynamics of single-dose oral tolvaptan in fasted and non-fasted states in healthy Caucasian and Japanese male subjects
    • S.E. Shoaf, S.R. Kim, P. Bricmont, and S. Mallikaarjun Pharmacokinetics and pharmacodynamics of single-dose oral tolvaptan in fasted and non-fasted states in healthy Caucasian and Japanese male subjects Eur. J. Clin. Pharmacol. 68 2012 1595 1603
    • (2012) Eur. J. Clin. Pharmacol. , vol.68 , pp. 1595-1603
    • Shoaf, S.E.1    Kim, S.R.2    Bricmont, P.3    Mallikaarjun, S.4
  • 62
    • 11144262703 scopus 로고    scopus 로고
    • Human variability in xenobiotic metabolism and pathway-related uncertainty factors for chemical risk assessment: A review
    • J.L.C.M. Dorne, K. Walton, and A.G. Renwick Human variability in xenobiotic metabolism and pathway-related uncertainty factors for chemical risk assessment: a review Food Chem. Toxicol. 43 2005 203 216
    • (2005) Food Chem. Toxicol. , vol.43 , pp. 203-216
    • Dorne, J.L.C.M.1    Walton, K.2    Renwick, A.G.3
  • 63
    • 0041342015 scopus 로고    scopus 로고
    • Drug-induced hepatotoxicity
    • W.M. Lee Drug-induced hepatotoxicity N. Engl. J. Med. 349 2003 474 485
    • (2003) N. Engl. J. Med. , vol.349 , pp. 474-485
    • Lee, W.M.1


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.