메뉴 건너뛰기




Volumn 13, Issue 30, 2007, Pages 3155-3167

Targeting of nuclear factor-κB and proteasome by dithiocarbamate complexes with metals

Author keywords

Diethyldithiocarbamate (DDTC); Disulfiram; JAMM; Metal dithiocarbamates; NF B; Proteasome; Pyrrolidinedithiocarbamate (PDTC)

Indexed keywords

ALDEHYDE DERIVATIVE; ANTIDOTE; ANTINEOPLASTIC AGENT; BETA LACTONE DERIVATIVE; BORONIC ACID DERIVATIVE; BORTEZOMIB; DIETHYLDITHIOCARBAMIC ACID; DISULFIRAM; DITHIOCARBAMIC ACID; DITHIOCARBAMIC ACID DERIVATIVE; EPOXYKETONE DERIVATIVE; ETHYLDITHIOCARBAMIC ACID; ETHYLMETHYLDITHIOCARBAMIC ACID; GLUCONATE ZINC; IMMUNOGLOBULIN ENHANCER BINDING PROTEIN; PESTICIDE; PROTEASOME; PROTEASOME INHIBITOR; PYRROLIDINEDITHIOCARBAMIC ACID; TRANSITION ELEMENT; UNCLASSIFIED DRUG; VINYL SULFONE DERIVATIVE; ZINC; ZINC DIBENZYLDITHIOCARBAMIC ACID;

EID: 36349012493     PISSN: 13816128     EISSN: None     Source Type: Journal    
DOI: 10.2174/138161207782110390     Document Type: Review
Times cited : (130)

References (344)
  • 2
    • 34648828162 scopus 로고
    • Electrochemistry and redox behavior of transition-metal dithiocarbamates
    • Bond AM, Martin RL. Electrochemistry and redox behavior of transition-metal dithiocarbamates. Coord Chem Rev 1984; 54: 23-98.
    • (1984) Coord Chem Rev , vol.54 , pp. 23-98
    • Bond, A.M.1    Martin, R.L.2
  • 3
    • 0030227105 scopus 로고    scopus 로고
    • Stereochemistry and molecular rearrangements of some six-, seven- mid eight-coordinate chelates of early transition metals
    • Fay RC. Stereochemistry and molecular rearrangements of some six-, seven- mid eight-coordinate chelates of early transition metals. Coord Chem Rev 1996; 154: 99-124.
    • (1996) Coord Chem Rev , vol.154 , pp. 99-124
    • Fay, R.C.1
  • 4
    • 0031494474 scopus 로고    scopus 로고
    • Photochemistry of Fe(III), Fe(IV), Ru(III), Mo(VI), and Ni(IV) dithiocarbamate complexes
    • Plyusnin VF, Grivin VP, Larionov SV. Photochemistry of Fe(III), Fe(IV), Ru(III), Mo(VI), and Ni(IV) dithiocarbamate complexes. Coord Chem Rev 1997; 159: 121-33.
    • (1997) Coord Chem Rev , vol.159 , pp. 121-133
    • Plyusnin, V.F.1    Grivin, V.P.2    Larionov, S.V.3
  • 5
    • 17644429135 scopus 로고    scopus 로고
    • Coordination chemistry of gold(II) complexes
    • Laguna A, Laguna M. Coordination chemistry of gold(II) complexes. Coord Chem Rev 1999; 193-5: 837-56.
    • (1999) Coord Chem Rev
    • Laguna, A.1    Laguna, M.2
  • 6
    • 0033826184 scopus 로고    scopus 로고
    • The reactivity of metal species towards thiuram sulfides: An alternative route to the syntheses of metal dithiocarbamates
    • Victoriano LI. The reactivity of metal species towards thiuram sulfides: an alternative route to the syntheses of metal dithiocarbamates. Coord Chem Rev 2000; 196: 383-98.
    • (2000) Coord Chem Rev , vol.196 , pp. 383-398
    • Victoriano, L.I.1
  • 8
    • 0037262749 scopus 로고    scopus 로고
    • Chemistry of arsenic, antimony and bismuth compounds derived from xanthate, dithiocarbamate and phosphoras based ligands
    • Garje SS, Jain VK. Chemistry of arsenic, antimony and bismuth compounds derived from xanthate, dithiocarbamate and phosphoras based ligands. Coord Chem Rev 2003; 236: 35-56.
    • (2003) Coord Chem Rev , vol.236 , pp. 35-56
    • Garje, S.S.1    Jain, V.K.2
  • 10
    • 20044363584 scopus 로고    scopus 로고
    • 13C CP/MAS NMR of dialkyldithiocarbamate compounds with Ni(II) and Zn(II)
    • 13C CP/MAS NMR of dialkyldithiocarbamate compounds with Ni(II) and Zn(II). Top Curr Chem 2005; 246: 271-337.
    • (2005) Top Curr Chem , vol.246 , pp. 271-337
    • Ivanov, A.V.1    Antzutkin, O.N.2
  • 11
    • 0001058533 scopus 로고    scopus 로고
    • Thiuramand dithiocarbamate-accelerated sulfur vulcanization from the chemist's perspective. Methods, materials and mechanisms reviewed
    • Nieuwenhuizen PJ, Reedijk J, van Duin M, McGill WJ. Thiuramand dithiocarbamate-accelerated sulfur vulcanization from the chemist's perspective. Methods, materials and mechanisms reviewed. Rubber Chem Technol 1997; 70: 368-429.
    • (1997) Rubber Chem Technol , vol.70 , pp. 368-429
    • Nieuwenhuizen, P.J.1    Reedijk, J.2    van Duin, M.3    McGill, W.J.4
  • 12
    • 0034300731 scopus 로고    scopus 로고
    • Effect of vulcanization systems and antioxidants on discoloration and degradation of natural rubber latex thread under UV radiation
    • Kurian JK, Peethambaran NR, Mary KC, Kuriakose B. Effect of vulcanization systems and antioxidants on discoloration and degradation of natural rubber latex thread under UV radiation. J Appl Polym Sci 2000; 78: 304-10.
    • (2000) J Appl Polym Sci , vol.78 , pp. 304-310
    • Kurian, J.K.1    Peethambaran, N.R.2    Mary, K.C.3    Kuriakose, B.4
  • 13
    • 0001247556 scopus 로고
    • Determination of copper as a dithiocarbamate complex by reversed-phase liquid-chromatography with electrochemical detection
    • Bond AM, Wallace GG. Determination of copper as a dithiocarbamate complex by reversed-phase liquid-chromatography with electrochemical detection. Anal Chem 1981; 53: 1209-13.
    • (1981) Anal Chem , vol.53 , pp. 1209-1213
    • Bond, A.M.1    Wallace, G.G.2
  • 14
    • 0020420495 scopus 로고
    • Solvent-extraction of dithiocarbamate complexes and back-extraction with mercury(II) for determination of trace-metals in sea-water by atomic-absorption spectrometry
    • Lo JM, Yu JC, Hutchison FI, Wal CM. Solvent-extraction of dithiocarbamate complexes and back-extraction with mercury(II) for determination of trace-metals in sea-water by atomic-absorption spectrometry. Anal Chem 1982; 54: 2536-9.
    • (1982) Anal Chem , vol.54 , pp. 2536-2539
    • JM, L.1    Yu, J.C.2    Hutchison, F.I.3    Wal, C.M.4
  • 15
    • 0000251467 scopus 로고    scopus 로고
    • Preconcentration of rare earth elements in seawater with poly(acrylaminophosphonicdithiocarbamate) chelating fiber prior to determination by inductively coupled plasma mass spectrometry
    • Zhang TH, Shan XQ, Liu RX, Tang HX, Zhang SZ. Preconcentration of rare earth elements in seawater with poly(acrylaminophosphonicdithiocarbamate) chelating fiber prior to determination by inductively coupled plasma mass spectrometry. Anal Chem 1998; 70: 3964-8.
    • (1998) Anal Chem , vol.70 , pp. 3964-3968
    • Zhang, T.H.1    Shan, X.Q.2    Liu, R.X.3    Tang, H.X.4    Zhang, S.Z.5
  • 16
    • 36348961620 scopus 로고    scopus 로고
    • Environmental Health Criteria, World Health Organization, Vammal, 1988.
    • Environmental Health Criteria, World Health Organization, Vammal, 1988.
  • 17
    • 0029550966 scopus 로고
    • International safety assessment of pesticides: Dithiocarbamate pesticides, ETU, and PTU - A review and update
    • Vettorazzi G, Almeida WF, Burin GJ, Jaeger RB, Puga FR, Rahde AF, et al. International safety assessment of pesticides: Dithiocarbamate pesticides, ETU, and PTU - A review and update. Yeratog Carcinog Mutagen 1995; 15: 313-37.
    • (1995) Yeratog Carcinog Mutagen , vol.15 , pp. 313-337
    • Vettorazzi, G.1    Almeida, W.F.2    Burin, G.J.3    Jaeger, R.B.4    Puga, F.R.5    Rahde, A.F.6
  • 18
    • 0032912534 scopus 로고    scopus 로고
    • Dithiocarbamate pesticides affect glutamate transport in brain synaptic vesicles
    • Vaccari A, Saba P, Mocci I, Ruiu S. Dithiocarbamate pesticides affect glutamate transport in brain synaptic vesicles. J Pharmacol Exp Ther 1999; 288: 1-5.
    • (1999) J Pharmacol Exp Ther , vol.288 , pp. 1-5
    • Vaccari, A.1    Saba, P.2    Mocci, I.3    Ruiu, S.4
  • 19
    • 0035175420 scopus 로고    scopus 로고
    • Effect of the dithiocarbamate pesticide zineb and its commercial formulation, azzurro. I. Genotoxic evaluation on cultured human lymphocytes exposed in vitro
    • Soloneski S, Gonzalez M, Piaggio E, Apezteguia M, Reigosa MA, Larramendy ML. Effect of the dithiocarbamate pesticide zineb and its commercial formulation, azzurro. I. Genotoxic evaluation on cultured human lymphocytes exposed in vitro. Mutagenesis 2001; 16: 487-93.
    • (2001) Mutagenesis , vol.16 , pp. 487-493
    • Soloneski, S.1    Gonzalez, M.2    Piaggio, E.3    Apezteguia, M.4    Reigosa, M.A.5    Larramendy, M.L.6
  • 20
    • 0036324009 scopus 로고    scopus 로고
    • Effect of the dithiocarbamate pesticide zineb and its commercial formulation, azzurro. II. Micronucleus induction in immunophenotyped human lymphocytes
    • Soloneski S, Reigosa MA, Larramendy ML. Effect of the dithiocarbamate pesticide zineb and its commercial formulation, azzurro. II. Micronucleus induction in immunophenotyped human lymphocytes. Environ Mol Mutagen 2002; 40: 57-62.
    • (2002) Environ Mol Mutagen , vol.40 , pp. 57-62
    • Soloneski, S.1    Reigosa, M.A.2    Larramendy, M.L.3
  • 21
    • 0037082996 scopus 로고    scopus 로고
    • Effect of the dithiocarbamate pesticide zineb and its commercial formulation, azzurro. III. Genotoxic evaluation on Chinese hamster ovary (CHO) cells
    • Soloneski S, Gonzalez M, Piaggio E, Reigosa MA, Larramendy ML. Effect of the dithiocarbamate pesticide zineb and its commercial formulation, azzurro. III. Genotoxic evaluation on Chinese hamster ovary (CHO) cells. Mutat Res 2002; 514: 201-12.
    • (2002) Mutat Res , vol.514 , pp. 201-212
    • Soloneski, S.1    Gonzalez, M.2    Piaggio, E.3    Reigosa, M.A.4    Larramendy, M.L.5
  • 22
    • 0037427913 scopus 로고    scopus 로고
    • Effect of the dithiocarbamate pesticide zineb and its commercial formulation, azzurro. IV. DNA damage and repair kinetics assessed by single cell gel electrophoresis (SCGE) assay on Chinese hamster ovary (CHO) cells
    • Gonzalez M, Soloneski S, Reigosa MA, Larramendy ML. Effect of the dithiocarbamate pesticide zineb and its commercial formulation, azzurro. IV. DNA damage and repair kinetics assessed by single cell gel electrophoresis (SCGE) assay on Chinese hamster ovary (CHO) cells. Murat Res 2003; 534: 145-54.
    • (2003) Murat Res , vol.534 , pp. 145-154
    • Gonzalez, M.1    Soloneski, S.2    Reigosa, M.A.3    Larramendy, M.L.4
  • 23
    • 0037457538 scopus 로고    scopus 로고
    • Effect of the dithiocarbamate pesticide zineb and its commercial formulation, azzurro. V. Abnormalities induced in the spindle apparatus of transformed and non-transformed mammalian cell lines
    • Soloneski S, Reigosa MA, Larramendy ML. Effect of the dithiocarbamate pesticide zineb and its commercial formulation, azzurro. V. Abnormalities induced in the spindle apparatus of transformed and non-transformed mammalian cell lines. Mutat Res 2003; 536: 121-9.
    • (2003) Mutat Res , vol.536 , pp. 121-129
    • Soloneski, S.1    Reigosa, M.A.2    Larramendy, M.L.3
  • 24
    • 0037837017 scopus 로고    scopus 로고
    • Abnormal fertilization is responsible for reduced fecundity following thiram-induced ovulatory delay in the rat
    • Stoker TE, Jeffay SC, Zucker RM, Cooper RL, Perreault SD. Abnormal fertilization is responsible for reduced fecundity following thiram-induced ovulatory delay in the rat. Biol Reprod 2003; 68: 2142-9.
    • (2003) Biol Reprod , vol.68 , pp. 2142-2149
    • Stoker, T.E.1    Jeffay, S.C.2    Zucker, R.M.3    Cooper, R.L.4    Perreault, S.D.5
  • 25
    • 32544449298 scopus 로고    scopus 로고
    • DNA damage and apoptosis induction by the pesticide mancozeb in rat cells: Involvement of the oxidative mechanism
    • Calviello G, Piccioni E, Boninsegna A, Tedesco B, Maggiano N, Serini S, et al. DNA damage and apoptosis induction by the pesticide mancozeb in rat cells: Involvement of the oxidative mechanism. Toxicol Appl Pharmacol 2006; 211: 87-96.
    • (2006) Toxicol Appl Pharmacol , vol.211 , pp. 87-96
    • Calviello, G.1    Piccioni, E.2    Boninsegna, A.3    Tedesco, B.4    Maggiano, N.5    Serini, S.6
  • 27
    • 0037265974 scopus 로고    scopus 로고
    • Manganese ethylene-bis-dithiocarbamate and selective dopaminergic neurodegeneration in rat: A link through mitochondrial dysfunction
    • Zhang J, Fitsanakis VA, Gu GY, Jing DQ, Ao MF, Amarnath V, et al. Manganese ethylene-bis-dithiocarbamate and selective dopaminergic neurodegeneration in rat: A link through mitochondrial dysfunction. J Neurochem 2003; 84: 336-46.
    • (2003) J Neurochem , vol.84 , pp. 336-346
    • Zhang, J.1    Fitsanakis, V.A.2    Gu, G.Y.3    Jing, D.Q.4    Ao, M.F.5    Amarnath, V.6
  • 28
    • 0038248977 scopus 로고    scopus 로고
    • Increased synaptosomal dopamine content and brain concentration of paraquat produced by selective. dithiocarbamates
    • Barlow BK, Thiruchelvam MJ, Bennice L, Cory-Slechta DA, Ballatori N, Richfield EK. Increased synaptosomal dopamine content and brain concentration of paraquat produced by selective. dithiocarbamates. J Neurochem 2003; 85: 1075-86.
    • (2003) J Neurochem , vol.85 , pp. 1075-1086
    • Barlow, B.K.1    Thiruchelvam, M.J.2    Bennice, L.3    Cory-Slechta, D.A.4    Ballatori, N.5    Richfield, E.K.6
  • 29
    • 5444255433 scopus 로고    scopus 로고
    • Neurotoxicant-induced animal models of Parkinson's disease: Understanding the role of rotenone, maneb and paraquat in neurodegeneration
    • Uversky VN. Neurotoxicant-induced animal models of Parkinson's disease: understanding the role of rotenone, maneb and paraquat in neurodegeneration. Cell Tissue Res 2004; 318: 225-41.
    • (2004) Cell Tissue Res , vol.318 , pp. 225-241
    • Uversky, V.N.1
  • 30
    • 27144556141 scopus 로고    scopus 로고
    • Evaluation of epidemiologic and animal data associating pesticides with Parkinson's disease
    • Li AA, Mink PJ, McIntosh LJ, Teta MJ, Finley B. Evaluation of epidemiologic and animal data associating pesticides with Parkinson's disease. J Occup Environ Med 2005; 47: 1059-87.
    • (2005) J Occup Environ Med , vol.47 , pp. 1059-1087
    • Li, A.A.1    Mink, P.J.2    McIntosh, L.J.3    Teta, M.J.4    Finley, B.5
  • 31
    • 20444471551 scopus 로고    scopus 로고
    • Overexpression of superoxide dismutase or glutathione peroxidase protects against the paraquat + maneb-induced Parkinson disease phenotype
    • Thiruchelvam M, Prokopenko O, Cory-Slechta DA, Richfield EK, Buckley A. Mirochnitchenko O. Overexpression of superoxide dismutase or glutathione peroxidase protects against the paraquat + maneb-induced Parkinson disease phenotype. J Biol Chem 2005; 280: 22530-9.
    • (2005) J Biol Chem , vol.280 , pp. 22530-22539
    • Thiruchelvam, M.1    Prokopenko, O.2    Cory-Slechta, D.A.3    Richfield, E.K.4    Buckley, A.5    Mirochnitchenko, O.6
  • 33
    • 0018425161 scopus 로고
    • Efficacy of sodium diethyldithiocabamate (dithiocarb) in acute nickel carbonyl poisoning
    • Sunderman FW. Efficacy of sodium diethyldithiocabamate (dithiocarb) in acute nickel carbonyl poisoning. Ann Clin Lab Sci 1979; 9: 1-10.
    • (1979) Ann Clin Lab Sci , vol.9 , pp. 1-10
    • Sunderman, F.W.1
  • 34
    • 0020964135 scopus 로고
    • Structure-activity-relationships among dithiocarbamate antidotes for acute cadmium chloride intoxication
    • Jone SG, Jones MM. Structure-activity-relationships among dithiocarbamate antidotes for acute cadmium chloride intoxication. Environ Health Perspect 1984; 54: 285-90.
    • (1984) Environ Health Perspect , vol.54 , pp. 285-290
    • Jone, S.G.1    Jones, M.M.2
  • 35
    • 0025887171 scopus 로고
    • Dithlocarbamate derivatives from α-amino acids as chelating agents for toxic metal ions
    • Macias B. Criado JJ, Vaquero MV, Villa MV, Castillo M. Dithlocarbamate derivatives from α-amino acids as chelating agents for toxic metal ions. J Inorg Biochem 1991;,42: 17-24.
    • (1991) J Inorg Biochem , vol.42 , pp. 17-24
    • Macias, B.1    Criado, J.J.2    Vaquero, M.V.3    Villa, M.V.4    Castillo, M.5
  • 36
    • 0025893056 scopus 로고
    • Chelation in metal intoxication. 36. Effect of substituted piperazine dithiocarbamates in lead exposed rats
    • Tandon SK, Kachru DN. Chelation in metal intoxication. 36. Effect of substituted piperazine dithiocarbamates in lead exposed rats. Acta Pharmacol Sin 1991; 12: 391-4.
    • (1991) Acta Pharmacol Sin , vol.12 , pp. 391-394
    • Tandon, S.K.1    Kachru, D.N.2
  • 37
    • 27744527460 scopus 로고    scopus 로고
    • Chelators as antidotes of metal toxicity: Therapeutic and experimental aspects
    • Blanusa M, Varnai VM, Piasek M, Kostial K. Chelators as antidotes of metal toxicity: Therapeutic and experimental aspects. Curr Med Chem 2005; 12: 2771-94.
    • (2005) Curr Med Chem , vol.12 , pp. 2771-2794
    • Blanusa, M.1    Varnai, V.M.2    Piasek, M.3    Kostial, K.4
  • 38
    • 0022542073 scopus 로고    scopus 로고
    • Effect of diethyldithiocarbamate on cis-diamminedicholoroplatinum(II)-induced cytotoxicity, DNA cross-linking, and gamma-glutamyltransferase transpeptidase inhibition
    • Bodenner DL, Dedon PC, Keng PC, Borch RF. Effect of diethyldithiocarbamate on cis-diamminedicholoroplatinum(II)-induced cytotoxicity, DNA cross-linking, and gamma-glutamyltransferase transpeptidase inhibition. Cancer Res 1986; 46: 2745-
    • Cancer Res 1986; 46: 2745
    • Bodenner, D.L.1    Dedon, P.C.2    Keng, P.C.3    Borch, R.F.4
  • 39
    • 0023732397 scopus 로고
    • High-dose carboplatin with diethyldithiocarbamate chemoprotection in treatment of women with relapsed ovarian cancer
    • Bothenberg ML, Ostchega Y, Steinberg SM, Young RC, Hummel S, Ozols RF. High-dose carboplatin with diethyldithiocarbamate chemoprotection in treatment of women with relapsed ovarian cancer. J Natl Cancer Inst 1988; 80: 1488-92.
    • (1988) J Natl Cancer Inst , vol.80 , pp. 1488-1492
    • Bothenberg, M.L.1    Ostchega, Y.2    Steinberg, S.M.3    Young, R.C.4    Hummel, S.5    Ozols, R.F.6
  • 40
  • 41
    • 33646428928 scopus 로고    scopus 로고
    • EXAFS and IR structural study of platinum-based anticancer drugs' degradation by diethyl dithiocarbamate
    • Bouvet D, Michalowicz A, Crauste-Manciet S, Brossard D, Provost K. EXAFS and IR structural study of platinum-based anticancer drugs' degradation by diethyl dithiocarbamate. Inorg Chem 2006; 45: 3393-8.
    • (2006) Inorg Chem , vol.45 , pp. 3393-3398
    • Bouvet, D.1    Michalowicz, A.2    Crauste-Manciet, S.3    Brossard, D.4    Provost, K.5
  • 42
    • 0028321926 scopus 로고
    • Spin trapping of nitric oxide produced in vivo in septic shock mice
    • Lai CS, Komarov AM. Spin trapping of nitric oxide produced in vivo in septic shock mice. FEBS Lett 1994; 345: 120-4.
    • (1994) FEBS Lett , vol.345 , pp. 120-124
    • Lai, C.S.1    Komarov, A.M.2
  • 43
    • 0028985256 scopus 로고
    • Direct measurement of nitric oxide generation in the ischemic heart using electron paramagnetic resonance spectroscopy
    • Zweier JL, Wang PH, Kuppusamy P. Direct measurement of nitric oxide generation in the ischemic heart using electron paramagnetic resonance spectroscopy. J Biol Chem 1995; 270: 304-7.
    • (1995) J Biol Chem , vol.270 , pp. 304-307
    • Zweier, J.L.1    Wang, P.H.2    Kuppusamy, P.3
  • 44
    • 0030840366 scopus 로고    scopus 로고
    • Direct measurement of nitric oxide generation from nitric oxide synthese
    • Xia Y, Zweier JL. Direct measurement of nitric oxide generation from nitric oxide synthese. Proc Natl Acad Sci USA 1997; 94: 12705-10.
    • (1997) Proc Natl Acad Sci USA , vol.94 , pp. 12705-12710
    • Xia, Y.1    Zweier, J.L.2
  • 45
    • 0032858864 scopus 로고    scopus 로고
    • Ex vivo EPR detection of nitric oxide in brain tissue
    • Fujii H, Berliner LJ. Ex vivo EPR detection of nitric oxide in brain tissue. Magn Reson Med 1999; 42: 599-602.
    • (1999) Magn Reson Med , vol.42 , pp. 599-602
    • Fujii, H.1    Berliner, L.J.2
  • 46
    • 0034695480 scopus 로고    scopus 로고
    • Nitric oxide forming reaction between the iron-N-methyl-D-glucamine dithiocarbamate complex and nitrite
    • Tsuchiya K, Yoshizumi M, Houchi H, Mason RP. Nitric oxide forming reaction between the iron-N-methyl-D-glucamine dithiocarbamate complex and nitrite. J Biol Chem 2000; 275: 1551-6.
    • (2000) J Biol Chem , vol.275 , pp. 1551-1556
    • Tsuchiya, K.1    Yoshizumi, M.2    Houchi, H.3    Mason, R.P.4
  • 47
    • 0033825916 scopus 로고    scopus 로고
    • A new trend in iron-dithiocarbamate complexes: As an endogenous NO trapping agent
    • Fujii S, Yoshimura T. A new trend in iron-dithiocarbamate complexes: As an endogenous NO trapping agent. Coord Chem Rev 2000; 198: 89-99.
    • (2000) Coord Chem Rev , vol.198 , pp. 89-99
    • Fujii, S.1    Yoshimura, T.2
  • 48
    • 0034667724 scopus 로고    scopus 로고
    • Electron paramagnetic resonance spectroscopy with N-methyl-D-glucamine dithiocarbamate iron complexes distinguishes nitric oxide and nitroxyl anion in a redox-dependent manner: Applications in identifying nitrogen monoxide products from nitric oxide synthese
    • Xia Y, Cardounel AJ, Vanin AF, Zweier JL. Electron paramagnetic resonance spectroscopy with N-methyl-D-glucamine dithiocarbamate iron complexes distinguishes nitric oxide and nitroxyl anion in a redox-dependent manner: Applications in identifying nitrogen monoxide products from nitric oxide synthese. Free Radic Biol Med 2000; 29: 793-7.
    • (2000) Free Radic Biol Med , vol.29 , pp. 793-797
    • Xia, Y.1    Cardounel, A.J.2    Vanin, A.F.3    Zweier, J.L.4
  • 49
    • 0036439013 scopus 로고    scopus 로고
    • Electron paramagnetic resonance for quantitation of nitric oxide in aqueous solutions
    • Venkataraman S, Martin SM, Buettner GR. Electron paramagnetic resonance for quantitation of nitric oxide in aqueous solutions. Methods Enzymol 2002; 359: 3-18.
    • (2002) Methods Enzymol , vol.359 , pp. 3-18
    • Venkataraman, S.1    Martin, S.M.2    Buettner, G.R.3
  • 50
    • 25444451350 scopus 로고    scopus 로고
    • Spin trapping nitric oxide from neuronal nitric oxide synthase: A look at several iron-dithiocarbamate complexes
    • Weaver J, Porasuphatana S, Tsai P, Budzichowski T, Rosen GM. Spin trapping nitric oxide from neuronal nitric oxide synthase: A look at several iron-dithiocarbamate complexes. Free Radic Res 2005; 39: 1027-33.
    • (2005) Free Radic Res , vol.39 , pp. 1027-1033
    • Weaver, J.1    Porasuphatana, S.2    Tsai, P.3    Budzichowski, T.4    Rosen, G.M.5
  • 52
    • 33751424161 scopus 로고    scopus 로고
    • Reduction enhances yields of nitric oxide trapping by iron-diethyldithiocarbamate complex in biological systems
    • Vanin AF, Bevers LM, Mikoyan VD, Poltorakov AP, Kubrina LN, van Faassen E. Reduction enhances yields of nitric oxide trapping by iron-diethyldithiocarbamate complex in biological systems. Nitric Oxide 2007; 16: 71-81.
    • (2007) Nitric Oxide , vol.16 , pp. 71-81
    • Vanin, A.F.1    Bevers, L.M.2    Mikoyan, V.D.3    Poltorakov, A.P.4    Kubrina, L.N.5    van Faassen, E.6
  • 53
    • 33947338778 scopus 로고
    • On the action of alkyl thiocyanates and alkyl isothiocyanates with thiol acids
    • Wheeler HL, Merriam HF. On the action of alkyl thiocyanates and alkyl isothiocyanates with thiol acids. J Am Chem Soc 1901; 23: 283-99.
    • (1901) J Am Chem Soc , vol.23 , pp. 283-299
    • Wheeler, H.L.1    Merriam, H.F.2
  • 54
    • 0242357460 scopus 로고
    • Metallic salts of dithiocarbamic acids; preparation of isothiocyanates in aliphatic series
    • Delepine M. Metallic salts of dithiocarbamic acids; preparation of isothiocyanates in aliphatic series. Compt Rend 1907; 144: 1125-7.
    • (1907) Compt Rend , vol.144 , pp. 1125-1127
    • Delepine, M.1
  • 55
    • 0038879900 scopus 로고
    • Reactions of accelerators during vulcanization
    • Bedford CW, Scott W. Reactions of accelerators during vulcanization. Ind Eng Chem 1920; 12: 31-3.
    • (1920) Ind Eng Chem , vol.12 , pp. 31-33
    • Bedford, C.W.1    Scott, W.2
  • 56
    • 0000416036 scopus 로고
    • Electronic structures of dithiocarbamates and xanthates
    • Chatt J, Duncanson LA, Venanzi LM. Electronic structures of dithiocarbamates and xanthates. Nature 1956; 177: 1042-3.
    • (1956) Nature , vol.177 , pp. 1042-1043
    • Chatt, J.1    Duncanson, L.A.2    Venanzi, L.M.3
  • 57
    • 0000725603 scopus 로고
    • Electron spin resonance and divalency of some dithiocarbamates of the coinage metals (Cu, Ag, Au)
    • Vanngard T, Akerstrom S. Electron spin resonance and divalency of some dithiocarbamates of the coinage metals (Cu, Ag, Au). Nature 1959; 184: 183-4.
    • (1959) Nature , vol.184 , pp. 183-184
    • Vanngard, T.1    Akerstrom, S.2
  • 58
    • 34250486810 scopus 로고
    • The properties of dithiocarbamates - a review
    • Halls DJ. The properties of dithiocarbamates - a review. Mikrochim Acta 1969; 62-77.
    • (1969) Mikrochim Acta , pp. 62-77
    • Halls, D.J.1
  • 59
    • 84981593751 scopus 로고
    • Reactions of complexes of dithiocarbamate and related ligands
    • Steggerda JJ, Cras JA, Willemse J. Reactions of complexes of dithiocarbamate and related ligands. Recl Trav Chim Pays Bas 1981; 100: 41-8.
    • (1981) Recl Trav Chim Pays Bas , vol.100 , pp. 41-48
    • Steggerda, J.J.1    Cras, J.A.2    Willemse, J.3
  • 60
    • 0023801173 scopus 로고
    • Optimal dithiocarbamate structure for immunomodulator action
    • Topping RJ, Jones MM. Optimal dithiocarbamate structure for immunomodulator action. Med Hypotheses 1988; 27: 55-7.
    • (1988) Med Hypotheses , vol.27 , pp. 55-57
    • Topping, R.J.1    Jones, M.M.2
  • 62
    • 0000925297 scopus 로고
    • A drug sensitizing the organism to ethyl alcohol
    • Hald J, Jacobsen E. A drug sensitizing the organism to ethyl alcohol. Lancet 1948; 252: 1001-4.
    • (1948) Lancet , vol.252 , pp. 1001-1004
    • Hald, J.1    Jacobsen, E.2
  • 63
    • 0030966158 scopus 로고    scopus 로고
    • The efficacy of disulfiram: A review of outcome studies
    • Hughes JC, Cook CCH. The efficacy of disulfiram: A review of outcome studies. Addiction 1997; 92: 381-95.
    • (1997) Addiction , vol.92 , pp. 381-395
    • Hughes, J.C.1    Cook, C.C.H.2
  • 65
    • 0033553203 scopus 로고    scopus 로고
    • Pharmacological treatment of alcohol dependence - a review of the evidence
    • Garbutt JC, West SL, Carey TS, Lohr KN, Crews FT. Pharmacological treatment of alcohol dependence - a review of the evidence. JAMA 1999; 281: 1318-25.
    • (1999) JAMA , vol.281 , pp. 1318-1325
    • Garbutt, J.C.1    West, S.L.2    Carey, T.S.3    Lohr, K.N.4    Crews, F.T.5
  • 66
    • 3042515206 scopus 로고    scopus 로고
    • Pharmacotherapy of alcohol dependence - a review of the clinical data
    • Mann K. Pharmacotherapy of alcohol dependence - a review of the clinical data. CNS Drugs 2004; 18: 485-504.
    • (2004) CNS Drugs , vol.18 , pp. 485-504
    • Mann, K.1
  • 67
    • 15544383967 scopus 로고    scopus 로고
    • Pharmacotherapy, pharmacogenomics, and the future of alcohol dependence treatment, part 1
    • Kenna GA, McGeary JE, Swift RM. Pharmacotherapy, pharmacogenomics, and the future of alcohol dependence treatment, part 1. Am J Health Syst Pharm 2004; 61: 2272-9.
    • (2004) Am J Health Syst Pharm , vol.61 , pp. 2272-2279
    • Kenna, G.A.1    McGeary, J.E.2    Swift, R.M.3
  • 68
    • 15544383967 scopus 로고    scopus 로고
    • Pharmacotherapy, pharmacogenomics, and the future of alcohol dependence treatment, part 2
    • Kenna GA, McGeary JE, Swift RM. Pharmacotherapy, pharmacogenomics, and the future of alcohol dependence treatment, part 2. Am J Health Syst Pharm 2004; 61: 2380-8.
    • (2004) Am J Health Syst Pharm , vol.61 , pp. 2380-2388
    • Kenna, G.A.1    McGeary, J.E.2    Swift, R.M.3
  • 70
    • 3242755117 scopus 로고    scopus 로고
    • Agents in development for the management of cocaine abuse
    • Gorelick DA, Gardner EL, Xi ZX. Agents in development for the management of cocaine abuse. Drugs 2004; 64: 1547-73.
    • (2004) Drugs , vol.64 , pp. 1547-1573
    • Gorelick, D.A.1    Gardner, E.L.2    Xi, Z.X.3
  • 71
    • 12844288905 scopus 로고    scopus 로고
    • Novel approaches to the treatment of cocaine addiction
    • Sofuoglu M, Kosten TR. Novel approaches to the treatment of cocaine addiction. CNS Drugs 2005; 19: 13-25.
    • (2005) CNS Drugs , vol.19 , pp. 13-25
    • Sofuoglu, M.1    Kosten, T.R.2
  • 73
    • 33749508432 scopus 로고    scopus 로고
    • New developments in the pharmacotherapy of cocaine dependence
    • Wiesbeck G, Duersteler-MacFarland K. New developments in the pharmacotherapy of cocaine dependence. Nervenarzt 2006; 77: 1064-70
    • (2006) Nervenarzt , vol.77 , pp. 1064-1070
    • Wiesbeck, G.1    Duersteler-MacFarland, K.2
  • 74
    • 0020033519 scopus 로고
    • Human aldehyde dehydrogenase: Mechanism of inhibition by disulfiram
    • Vallari, RC, Pietruszko R. Human aldehyde dehydrogenase: Mechanism of inhibition by disulfiram. Science 1982; 216: 637-9.
    • (1982) Science , vol.216 , pp. 637-639
    • Vallari, R.C.1    Pietruszko, R.2
  • 75
    • 0020504341 scopus 로고
    • Mechanism of inactivation of sheep liver cytoplasmic aldehyde dehydrogenase by disulfiram
    • Kitson TM. Mechanism of inactivation of sheep liver cytoplasmic aldehyde dehydrogenase by disulfiram. Biochem J 1983; 213: 551-4.
    • (1983) Biochem J , vol.213 , pp. 551-554
    • Kitson, T.M.1
  • 76
    • 0023156479 scopus 로고
    • Inactivation of horse liver mitochondrial aldehyde dehydrogenase by disulfiram
    • Sanny CG, Weiner H. Inactivation of horse liver mitochondrial aldehyde dehydrogenase by disulfiram. Biochem J 1987; 242: 499-503.
    • (1987) Biochem J , vol.242 , pp. 499-503
    • Sanny, C.G.1    Weiner, H.2
  • 77
    • 0034308085 scopus 로고    scopus 로고
    • Role of disulfiram in the in vitro inhibition of rat liver mitochondrial aldehyde dehydrogenase
    • Shen ML, Lipsky JJ, Naylor S. Role of disulfiram in the in vitro inhibition of rat liver mitochondrial aldehyde dehydrogenase. Biochem Pharmacol 2000; 60: 947-53.
    • (2000) Biochem Pharmacol , vol.60 , pp. 947-953
    • Shen, M.L.1    Lipsky, J.J.2    Naylor, S.3
  • 78
    • 0024580954 scopus 로고
    • Diethylthiocarbamic acid methyl ester: A potent inhibitor of aldehyde dehydrogenase found in rats treated with disulfiram or diethyldithiocarbamic acid methyl ester
    • Johansson B, Petersen EN, Arnold E. Diethylthiocarbamic acid methyl ester: A potent inhibitor of aldehyde dehydrogenase found in rats treated with disulfiram or diethyldithiocarbamic acid methyl ester. Biochem Pharmacol 1989; 38: 1053-9.
    • (1989) Biochem Pharmacol , vol.38 , pp. 1053-1059
    • Johansson, B.1    Petersen, E.N.2    Arnold, E.3
  • 79
    • 0024322043 scopus 로고
    • Inhibition of human erythrocyte and leukocyte aldehyde dehydrogenase activities by diethylthiocarbamic acid methyl ester - an in vivo metabolite of disulfiram
    • Helander A, Johansson B. Inhibition of human erythrocyte and leukocyte aldehyde dehydrogenase activities by diethylthiocarbamic acid methyl ester - an in vivo metabolite of disulfiram. Biochem Pharmacol 1989; 38: 2195-8.
    • (1989) Biochem Pharmacol , vol.38 , pp. 2195-2198
    • Helander, A.1    Johansson, B.2
  • 80
    • 0024325889 scopus 로고
    • Inhibition of erythrocyte aldehyde dehydrogenase activity and elimination kinetics of diethyldithiocarbamic acid methyl ester and its monothio analog after administration of single and repeated doses of disulfiram to man
    • Johansson B, Stankiewicz Z. Inhibition of erythrocyte aldehyde dehydrogenase activity and elimination kinetics of diethyldithiocarbamic acid methyl ester and its monothio analog after administration of single and repeated doses of disulfiram to man. Eur J Clin Pharmacol 1989; 37: 133-8.
    • (1989) Eur J Clin Pharmacol , vol.37 , pp. 133-138
    • Johansson, B.1    Stankiewicz, Z.2
  • 82
    • 0026439016 scopus 로고
    • A review of the pharmacokinetics and pharmacodynamics of disulfiram and its metabolites
    • Johansson B. A review of the pharmacokinetics and pharmacodynamics of disulfiram and its metabolites. Acta Psychiatr Scand 1992; 86: 15-26.
    • (1992) Acta Psychiatr Scand , vol.86 , pp. 15-26
    • Johansson, B.1
  • 83
    • 0028070184 scopus 로고
    • Identification of novel glutathione conjugates of disulfiram and diethyldithiocarbamate in rat bile by liquid chromatography tandem mass spectrometry - evidence for metabolic activation of disulfiram in vivo
    • Jin LX, Davis MR, Hu P, Baillie TA. Identification of novel glutathione conjugates of disulfiram and diethyldithiocarbamate in rat bile by liquid chromatography tandem mass spectrometry - evidence for metabolic activation of disulfiram in vivo. Chem Res Toxicol 1994; 7: 526-33.
    • (1994) Chem Res Toxicol , vol.7 , pp. 526-533
    • Jin, L.X.1    Davis, M.R.2    Hu, P.3    Baillie, T.A.4
  • 84
    • 0031581881 scopus 로고    scopus 로고
    • Inhibition of aldehyde dehydrogenase by disulfiram and its metabolite methyl diethylthiocarbamoylsulfoxide
    • Veverka KA, Johnson KL, Mays DC, Lipsky JJ, Naylor S. Inhibition of aldehyde dehydrogenase by disulfiram and its metabolite methyl diethylthiocarbamoylsulfoxide. Biochem Pharmacol 1997; 53: 511-8.
    • (1997) Biochem Pharmacol , vol.53 , pp. 511-518
    • Veverka, K.A.1    Johnson, K.L.2    Mays, D.C.3    Lipsky, J.J.4    Naylor, S.5
  • 85
    • 0027135997 scopus 로고
    • Bioactivation of S-methyl-N,N-diethylthiolcarbamate to S-methyl-N,N-diethylthiolcarbamate sulfoxide - implications for the role of cytochrome P450
    • Hart BW, Faiman MD. Bioactivation of S-methyl-N,N-diethylthiolcarbamate to S-methyl-N,N-diethylthiolcarbamate sulfoxide - implications for the role of cytochrome P450. Biochem Pharmacol 1993; 46: 2285-90.
    • (1993) Biochem Pharmacol , vol.46 , pp. 2285-2290
    • Hart, B.W.1    Faiman, M.D.2
  • 86
    • 0027131401 scopus 로고
    • Role of flavin-dependent monooxygenases and cytochrome P450 enzymes in the sulfoxidation of S-methyl-N,N-diethylthiolcarbamate
    • Madan A, Parkinson A, Faiman MD. Role of flavin-dependent monooxygenases and cytochrome P450 enzymes in the sulfoxidation of S-methyl-N,N-diethylthiolcarbamate. Biochem Pharmacol 1993; 46: 2291-7.
    • (1993) Biochem Pharmacol , vol.46 , pp. 2291-2297
    • Madan, A.1    Parkinson, A.2    Faiman, M.D.3
  • 87
    • 0035283181 scopus 로고    scopus 로고
    • Determination of in vivo adducts of disulfiram with mitochondrial aldehyde dehydrogenase
    • Shen ML, Johnson KL, Mays DC, Lipsky JJ, Naylor S. Determination of in vivo adducts of disulfiram with mitochondrial aldehyde dehydrogenase. Biochem Pharmacol 2001; 61: 537-45.
    • (2001) Biochem Pharmacol , vol.61 , pp. 537-545
    • Shen, M.L.1    Johnson, K.L.2    Mays, D.C.3    Lipsky, J.J.4    Naylor, S.5
  • 88
    • 0035969887 scopus 로고    scopus 로고
    • Overview - in vitro inhibition of aldehyde dehydrogenase by disulfiram and metabolites
    • Lipsky JL, Shen ML, Naylor S. Overview - in vitro inhibition of aldehyde dehydrogenase by disulfiram and metabolites. Chem Biol Interact 2001; 130: 81-91.
    • (2001) Chem Biol Interact , vol.130 , pp. 81-91
    • Lipsky, J.L.1    Shen, M.L.2    Naylor, S.3
  • 89
    • 0027196909 scopus 로고
    • Ditiocarb: Decomposition in aqueous solution and effect of the volatile products on its pharmacological use
    • Martens T, Langevin-Bermond D, Fleury MB. Ditiocarb: Decomposition in aqueous solution and effect of the volatile products on its pharmacological use. J Pharm Sci 1993; 82: 379-83.
    • (1993) J Pharm Sci , vol.82 , pp. 379-383
    • Martens, T.1    Langevin-Bermond, D.2    Fleury, M.B.3
  • 91
    • 0014028235 scopus 로고
    • Interaction between tetraethylthiuram disulflde and sulfhydryl groups of D-aminoacid oxidase and of hemoglobin
    • Neims AH, Coffey DS, Hellerma L. Interaction between tetraethylthiuram disulflde and sulfhydryl groups of D-aminoacid oxidase and of hemoglobin. J Biol Chem 1966; 241: 5941-8.
    • (1966) J Biol Chem , vol.241 , pp. 5941-5948
    • Neims, A.H.1    Coffey, D.S.2    Hellerma, L.3
  • 92
    • 0022481187 scopus 로고
    • A novel interaction of diethyldithiocarbamate with glutathione glutathione-peroxidase system
    • Kumar KS, Sancho AM, Weiss JF. A novel interaction of diethyldithiocarbamate with glutathione glutathione-peroxidase system. Int J Radiat Oncol Biol Phys 1986; 12: 1463-7.
    • (1986) Int J Radiat Oncol Biol Phys , vol.12 , pp. 1463-1467
    • Kumar, K.S.1    Sancho, A.M.2    Weiss, J.F.3
  • 93
    • 0026583741 scopus 로고
    • Diethyldithiocarbamate (ditiocarb sodium) effect on arachidonic acid metabolism in human mononuclear cells - glutathione-peroxidase-like activity
    • Hosni M, Meskini N, Prigent AF, Anker G, Joulain C, Elhabib R, et al. Diethyldithiocarbamate (ditiocarb sodium) effect on arachidonic acid metabolism in human mononuclear cells - glutathione-peroxidase-like activity. Biochem Pharmacol 1992; 43: 1319-29.
    • (1992) Biochem Pharmacol , vol.43 , pp. 1319-1329
    • Hosni, M.1    Meskini, N.2    Prigent, A.F.3    Anker, G.4    Joulain, C.5    Elhabib, R.6
  • 94
    • 0018778879 scopus 로고
    • In vivo and in vitro effects of thiuram disulfides and dithiocarbamates on hepatic microsomal drug metabolism in the rat
    • Zemaitis MA, Greene FE. In vivo and in vitro effects of thiuram disulfides and dithiocarbamates on hepatic microsomal drug metabolism in the rat. Toxicol Appl Pharmacol 1979; 48: 343-50.
    • (1979) Toxicol Appl Pharmacol , vol.48 , pp. 343-350
    • Zemaitis, M.A.1    Greene, F.E.2
  • 95
    • 0021338892 scopus 로고
    • In vitro interaction of dithiocarb with rat liver glutathione S-transferases
    • Dierickx PJ. In vitro interaction of dithiocarb with rat liver glutathione S-transferases. Pharmacol Res Commun 1984; 16: 135-43.
    • (1984) Pharmacol Res Commun , vol.16 , pp. 135-143
    • Dierickx, P.J.1
  • 96
    • 0023001898 scopus 로고
    • Inhibition of erythrocyte superoxide dismutase by diethyldithiocarbamate also results in oxyhemoglobin-catalyzed glutathione depletion and methemoglobin production
    • Kelner MJ, Alexander NM. Inhibition of erythrocyte superoxide dismutase by diethyldithiocarbamate also results in oxyhemoglobin-catalyzed glutathione depletion and methemoglobin production. J Biol Chem 1986; 261: 1636-41.
    • (1986) J Biol Chem , vol.261 , pp. 1636-1641
    • Kelner, M.J.1    Alexander, N.M.2
  • 97
    • 0024515189 scopus 로고
    • Inactivation of intracellular copper-zinc superoxide dismutase by copper chelating agents without glutathione depletion and methemoglobin formation
    • Kelner MJ, Bagnell R, Hale B, Alexander NM. Inactivation of intracellular copper-zinc superoxide dismutase by copper chelating agents without glutathione depletion and methemoglobin formation. Free Radic Biol Med 1989; 6: 355-60.
    • (1989) Free Radic Biol Med , vol.6 , pp. 355-360
    • Kelner, M.J.1    Bagnell, R.2    Hale, B.3    Alexander, N.M.4
  • 98
    • 0021358608 scopus 로고
    • Kinetics of the histological changes in lymphoid organs and of the T-cell inducing capacity of serum in mice treated with imuthiol (sodium diethyldithiocarbamate)
    • Pompidou A, Renoux M, Guillaumin JM, Mace B, Michel P, Coutance F, et al. Kinetics of the histological changes in lymphoid organs and of the T-cell inducing capacity of serum in mice treated with imuthiol (sodium diethyldithiocarbamate). Int Arch Allergy Appl Immunol 1984; 74: 172-7.
    • (1984) Int Arch Allergy Appl Immunol , vol.74 , pp. 172-177
    • Pompidou, A.1    Renoux, M.2    Guillaumin, J.M.3    Mace, B.4    Michel, P.5    Coutance, F.6
  • 99
    • 0021971823 scopus 로고
    • The generation and regulation of human lymphocytes T by imuthiol - evidence from an in vitro differentiation induction system
    • Pompidou A, Duchet N, Cooper MD, Mace B, Telvi L, Coutance F, et al. The generation and regulation of human lymphocytes T by imuthiol - evidence from an in vitro differentiation induction system. Int J Immunopharmacol 1985; 7: 561-6.
    • (1985) Int J Immunopharmacol , vol.7 , pp. 561-566
    • Pompidou, A.1    Duchet, N.2    Cooper, M.D.3    Mace, B.4    Telvi, L.5    Coutance, F.6
  • 100
    • 0022370567 scopus 로고
    • Isoprinosine and imuthiol, two potentially active compounds in patients with AIDS-related complex symptoms
    • Pompidou A, Delsaux MC, Telvi L, Mace B, Coutance F, Falkenrodt A, et al. Isoprinosine and imuthiol, two potentially active compounds in patients with AIDS-related complex symptoms. Cancer Res 1985; 45: 4671-3.
    • (1985) Cancer Res , vol.45 , pp. 4671-4673
    • Pompidou, A.1    Delsaux, M.C.2    Telvi, L.3    Mace, B.4    Coutance, F.5    Falkenrodt, A.6
  • 102
    • 0023679964 scopus 로고
    • Randomized, double blind, placebo controlled trial of ditiocarb sodium (imuthiol) in human immunodeficiciency virus infection
    • Lang JM, Trepo C, Kirstetter M, Herviou L, Retornaz G, Renoux G, et al. Randomized, double blind, placebo controlled trial of ditiocarb sodium (imuthiol) in human immunodeficiciency virus infection. Lancet 1988; 2: 702-6.
    • (1988) Lancet , vol.2 , pp. 702-706
    • Lang, J.M.1    Trepo, C.2    Kirstetter, M.3    Herviou, L.4    Retornaz, G.5    Renoux, G.6
  • 103
    • 0025598869 scopus 로고
    • Alternative therapies in HIV infection
    • Abrams DI. Alternative therapies in HIV infection. AIDS 1990; 4: 1179-87.
    • (1990) AIDS , vol.4 , pp. 1179-1187
    • Abrams, D.I.1
  • 104
    • 0025959545 scopus 로고
    • Therapeutic properties of sodium diethyldithiocarbamate - its role as an inhibitor in the progression of AIDS
    • Sunderman FW. Therapeutic properties of sodium diethyldithiocarbamate - its role as an inhibitor in the progression of AIDS. Ann Clin Lab Sci 1991; 21: 70-81.
    • (1991) Ann Clin Lab Sci , vol.21 , pp. 70-81
    • Sunderman, F.W.1
  • 105
    • 0025844784 scopus 로고
    • Ditiocarb sodium (diethyldithiocarbamate) therapy in patients with symptomatic HIV infection and AIDS. A randomized, double-blind, placebo controlled, multicenter study
    • Hersh EM, Brewton G, Abrams D, Bartlett J, Galpin J, Gill P, et al. Ditiocarb sodium (diethyldithiocarbamate) therapy in patients with symptomatic HIV infection and AIDS. A randomized, double-blind, placebo controlled, multicenter study. JAMA 1991; 265: 1538-44.
    • (1991) JAMA , vol.265 , pp. 1538-1544
    • Hersh, E.M.1    Brewton, G.2    Abrams, D.3    Bartlett, J.4    Galpin, J.5    Gill, P.6
  • 106
    • 0027402615 scopus 로고
    • Multicenter, randomized, placebo-controlled study of ditiocarb (imuthiol) in human immunodeficiency virus-infected asymptomatic and minimally symptomatic patients
    • Picolet H, Lang JM, Touraine JL, Livrozet JM, Saintmarc T, Kirstetter M, et al. Multicenter, randomized, placebo-controlled study of ditiocarb (imuthiol) in human immunodeficiency virus-infected asymptomatic and minimally symptomatic patients. AIDS Res Hum Retroviruses 1993; 9: 83-9.
    • (1993) AIDS Res Hum Retroviruses , vol.9 , pp. 83-89
    • Picolet, H.1    Lang, J.M.2    Touraine, J.L.3    Livrozet, J.M.4    Saintmarc, T.5    Kirstetter, M.6
  • 108
    • 0018604230 scopus 로고
    • Reaction of copper-zinc superoxide dismutase with diethylithiocarbamate
    • Misra HP. Reaction of copper-zinc superoxide dismutase with diethylithiocarbamate. J Biol Chem 1979; 254: 11623-8.
    • (1979) J Biol Chem , vol.254 , pp. 11623-11628
    • Misra, H.P.1
  • 109
    • 0019877295 scopus 로고
    • Reexamination of the reaction of diethyldithiocarbamate with the copper of superoxide dismutase
    • Cocco D, Calabrese L, Rigo A, Argese E, Rotilio G. Reexamination of the reaction of diethyldithiocarbamate with the copper of superoxide dismutase. J Biol Chem 1981; 256: 8983-6.
    • (1981) J Biol Chem , vol.256 , pp. 8983-8986
    • Cocco, D.1    Calabrese, L.2    Rigo, A.3    Argese, E.4    Rotilio, G.5
  • 110
    • 0000846251 scopus 로고
    • Structural studies of metal dithiocarbamates. II. The crystal and molecular structure of copper diethyldithiocarbamate
    • Bonamico M, Dessy G, Mugnoli A, Vaciago X, Zambonelli L. Structural studies of metal dithiocarbamates. II. The crystal and molecular structure of copper diethyldithiocarbamate. Acta Crystallogr 1965; 19: 886-97.
    • (1965) Acta Crystallogr , vol.19 , pp. 886-897
    • Bonamico, M.1    Dessy, G.2    Mugnoli, A.3    Vaciago, X.4    Zambonelli, L.5
  • 111
    • 0019754216 scopus 로고
    • Preparation of selectively metal free and metal substituted derivatives by reaction of Cu-Zn superoxide dismutase with diethyldithiocarbamate
    • Cocco D, Calabrese L, Rigo A, Marmocchi F, Rotilio G. Preparation of selectively metal free and metal substituted derivatives by reaction of Cu-Zn superoxide dismutase with diethyldithiocarbamate. Biochem J 1981; 199: 675-80.
    • (1981) Biochem J , vol.199 , pp. 675-680
    • Cocco, D.1    Calabrese, L.2    Rigo, A.3    Marmocchi, F.4    Rotilio, G.5
  • 112
    • 0017294656 scopus 로고
    • In vivo inhibition of superoxide dismutase in mice by diethyldithiocarbamate
    • Heikkila RE, Cabbat FS, Cohen G. In vivo inhibition of superoxide dismutase in mice by diethyldithiocarbamate. J Biol Chem 1976; 251: 2182-5.
    • (1976) J Biol Chem , vol.251 , pp. 2182-2185
    • Heikkila, R.E.1    Cabbat, F.S.2    Cohen, G.3
  • 113
    • 0031588551 scopus 로고    scopus 로고
    • Peroxidative activity of human Cu,Zn-superoxide dismutase
    • Kim SM, Kang JH. Peroxidative activity of human Cu,Zn-superoxide dismutase. Mol Cells 1997; 7: 120-4.
    • (1997) Mol Cells , vol.7 , pp. 120-124
    • Kim, S.M.1    Kang, J.H.2
  • 114
    • 0025992864 scopus 로고
    • Oxidation of toxic and carcinogenic chemicals by human cytochrome P450 enzymes
    • Guengerich FP, Shimada T. Oxidation of toxic and carcinogenic chemicals by human cytochrome P450 enzymes. Chem Res Toxicol 1991; 4: 391-407.
    • (1991) Chem Res Toxicol , vol.4 , pp. 391-407
    • Guengerich, F.P.1    Shimada, T.2
  • 115
    • 0019835546 scopus 로고
    • Inhibitors of cytochrome P-450s and their mechanism of action
    • Testa B, Jenner P. Inhibitors of cytochrome P-450s and their mechanism of action. Drug Metab Rev 1981; 12: 1-117.
    • (1981) Drug Metab Rev , vol.12 , pp. 1-117
    • Testa, B.1    Jenner, P.2
  • 116
    • 0028174881 scopus 로고
    • Evaluation of triacetylole-andomycin, α-naftoflavone and diethyldithiocarbamate as selective chemical probes for inhibition of human cytochromes P450
    • Chang TKH, Gonzalez FJ, Waxman DJ. Evaluation of triacetylole-andomycin, α-naftoflavone and diethyldithiocarbamate as selective chemical probes for inhibition of human cytochromes P450. Arch Biochem Biophys 1994; 311: 437-42.
    • (1994) Arch Biochem Biophys , vol.311 , pp. 437-442
    • Chang, T.K.H.1    Gonzalez, F.J.2    Waxman, D.J.3
  • 117
    • 0028858960 scopus 로고
    • Cytochrome P450 inhibitors - evaluation of specificities in the in vitro metabolism of therapeutic agents by human liver microsomes
    • Newton DJ, Wang RW, Lu AYH. Cytochrome P450 inhibitors - evaluation of specificities in the in vitro metabolism of therapeutic agents by human liver microsomes. Drug Metab Dispos 1995; 23: 154-8.
    • (1995) Drug Metab Dispos , vol.23 , pp. 154-158
    • Newton, D.J.1    Wang, R.W.2    Lu, A.Y.H.3
  • 118
    • 0030056594 scopus 로고    scopus 로고
    • Specificity of substrate and inhibitor probes for cytochrome P450s: Evaluation of in vitro metabolism using cDNA-expressed human P450s and human liver microsomes
    • Ono S, Hatanaka T, Hotta H, Satoh T, Gonzalez FJ, Tsutsui M. Specificity of substrate and inhibitor probes for cytochrome P450s: Evaluation of in vitro metabolism using cDNA-expressed human P450s and human liver microsomes. Xenobiotica 1996; 26: 681-93.
    • (1996) Xenobiotica , vol.26 , pp. 681-693
    • Ono, S.1    Hatanaka, T.2    Hotta, H.3    Satoh, T.4    Gonzalez, F.J.5    Tsutsui, M.6
  • 122
    • 0028026741 scopus 로고
    • A role for metals and free radicals in the induction of apoptosis in thymocytes
    • Wolfe JT, Ross D, Cohen GM. A role for metals and free radicals in the induction of apoptosis in thymocytes. FEBS Lett 1994; 352: 58-62.
    • (1994) FEBS Lett , vol.352 , pp. 58-62
    • Wolfe, J.T.1    Ross, D.2    Cohen, G.M.3
  • 124
    • 0343376119 scopus 로고    scopus 로고
    • Mechanism of dithiocarbamate inhibition of apoptosis: Thiol oxidation by dithiocarbamate disulfides directly inhibits processing of the caspase-3 proenzyme
    • Nobel CSI, Burgess DH, Zhivotovsky B, Burkitt MJ, Orenius S, Slater AFG. Mechanism of dithiocarbamate inhibition of apoptosis: Thiol oxidation by dithiocarbamate disulfides directly inhibits processing of the caspase-3 proenzyme. Chem Res Toxicol 1997; 10: 636-43.
    • (1997) Chem Res Toxicol , vol.10 , pp. 636-643
    • Nobel, C.S.I.1    Burgess, D.H.2    Zhivotovsky, B.3    Burkitt, M.J.4    Orenius, S.5    Slater, A.F.G.6
  • 126
    • 0028815621 scopus 로고
    • Dithiocarbamates induce apoptosis in thymocytes by raising the intracellular level of redox-active copper
    • Nobel CSI, Kimland M, Lind B, Orrenius S, Slater AFG. Dithiocarbamates induce apoptosis in thymocytes by raising the intracellular level of redox-active copper. J Biol Chem 1995; 270: 26202-8.
    • (1995) J Biol Chem , vol.270 , pp. 26202-26208
    • Nobel, C.S.I.1    Kimland, M.2    Lind, B.3    Orrenius, S.4    Slater, A.F.G.5
  • 127
    • 0031825806 scopus 로고    scopus 로고
    • Dithiocarbamate toxicity toward thymocytes involves their copper-catalyzed conversion to thiuram disulfides, which oxidize glutathione in a redox cycle without the release of reactive oxygen species
    • Burkitt MJ, Bishop HS, Milne L, Tsang SY, Provan GJ, Nobel CSI, et al. Dithiocarbamate toxicity toward thymocytes involves their copper-catalyzed conversion to thiuram disulfides, which oxidize glutathione in a redox cycle without the release of reactive oxygen species. Arch Biochem Biophys 1998; 353: 73-84.
    • (1998) Arch Biochem Biophys , vol.353 , pp. 73-84
    • Burkitt, M.J.1    Bishop, H.S.2    Milne, L.3    Tsang, S.Y.4    Provan, G.J.5    Nobel, C.S.I.6
  • 128
    • 0018758689 scopus 로고
    • Increased brain uptake of copper and zinc in mice caused by diethyldithiocarbamate
    • Aaseth J, Soli NE, Forre O. Increased brain uptake of copper and zinc in mice caused by diethyldithiocarbamate. Acta Pharmacol Toxicol 1979; 45: 41-4.
    • (1979) Acta Pharmacol Toxicol , vol.45 , pp. 41-44
    • Aaseth, J.1    Soli, N.E.2    Forre, O.3
  • 129
    • 0023951787 scopus 로고
    • Protective effects of glutathione on diethyldithiocarbamate (DDC) cyto-toxicity: A possible mechanism
    • Trombetta LD, Toulon M, Jamall IS. Protective effects of glutathione on diethyldithiocarbamate (DDC) cyto-toxicity: A possible mechanism. Toxicol Appl Pharmacol 1988; 93: 154-64.
    • (1988) Toxicol Appl Pharmacol , vol.93 , pp. 154-164
    • Trombetta, L.D.1    Toulon, M.2    Jamall, I.S.3
  • 130
    • 0034522975 scopus 로고    scopus 로고
    • Regulation of thyroid follicular cell function by intracellular redox-active copper
    • Iseki A, Kambe F, Okumura K, Hayakawa T, Seo H. Regulation of thyroid follicular cell function by intracellular redox-active copper. Endocrinology 2000; 141: 4373-82.
    • (2000) Endocrinology , vol.141 , pp. 4373-4382
    • Iseki, A.1    Kambe, F.2    Okumura, K.3    Hayakawa, T.4    Seo, H.5
  • 131
    • 0026067239 scopus 로고
    • Diethyldithiocarbamate, copper and neurological disorders
    • Allain P, Krari N. Diethyldithiocarbamate, copper and neurological disorders. Life Sci 1991; 48: 291-9.
    • (1991) Life Sci , vol.48 , pp. 291-299
    • Allain, P.1    Krari, N.2
  • 133
    • 0033799739 scopus 로고    scopus 로고
    • Disulfirain produces a non-carbon-disulfide-dependent Schwannopathy in the rat
    • Tonkin EG, Erve JCL, Valentine WM. Disulfirain produces a non-carbon-disulfide-dependent Schwannopathy in the rat. J Neuropathol Exp Neurol 2000; 59: 786-97.
    • (2000) J Neuropathol Exp Neurol , vol.59 , pp. 786-797
    • Tonkin, E.G.1    Erve, J.C.L.2    Valentine, W.M.3
  • 134
    • 4444223239 scopus 로고    scopus 로고
    • N,N-diethyldithiocarbamate produces copper accumulation, lipid peroxidation, and myelin injury in rat peripheral nerve
    • Tonkin EG, Valentine HL, Milatovic DM, Valentine WM. N,N-diethyldithiocarbamate produces copper accumulation, lipid peroxidation, and myelin injury in rat peripheral nerve. Toxicol Sci 2004; 81: 160-71.
    • (2004) Toxicol Sci , vol.81 , pp. 160-171
    • Tonkin, E.G.1    Valentine, H.L.2    Milatovic, D.M.3    Valentine, W.M.4
  • 135
    • 21344435513 scopus 로고    scopus 로고
    • Repeated exposure to pyrrolidine-dithiocarbamate induces peripheral nerve alterations in rats
    • Calviello G, Filippi GM, Toesca A, Palozza P, Maggiano N, Di Nicuolo F, et al. Repeated exposure to pyrrolidine-dithiocarbamate induces peripheral nerve alterations in rats. Toxicol Lett 2005; 158: 61-71
    • (2005) Toxicol Lett , vol.158 , pp. 61-71
    • Calviello, G.1    Filippi, G.M.2    Toesca, A.3    Palozza, P.4    Maggiano, N.5    Di Nicuolo, F.6
  • 136
    • 31144443476 scopus 로고    scopus 로고
    • Dietary copper enhances the peripheral myelinopathy produced by oral pyrrolidine dithiocarbamate
    • Valentine HL, Amarnath K, Amarnath V, Valentine WM. Dietary copper enhances the peripheral myelinopathy produced by oral pyrrolidine dithiocarbamate. Toxicol Sci 2006; 89: 485-94.
    • (2006) Toxicol Sci , vol.89 , pp. 485-494
    • Valentine, H.L.1    Amarnath, K.2    Amarnath, V.3    Valentine, W.M.4
  • 138
    • 0000852065 scopus 로고    scopus 로고
    • Pyrrolidine dithiocarbamate induces bovine cerebral endothelial cell death by increasing the intracellular zinc level
    • Kim CH, Kim JH, Xu J, Hsu CY, Ahn YS. Pyrrolidine dithiocarbamate induces bovine cerebral endothelial cell death by increasing the intracellular zinc level. J Neurochem 1999; 72: 1586-92.
    • (1999) J Neurochem , vol.72 , pp. 1586-1592
    • Kim, C.H.1    Kim, J.H.2    Xu, J.3    Hsu, C.Y.4    Ahn, Y.S.5
  • 139
    • 0037369564 scopus 로고    scopus 로고
    • Pyrrolidine dithiocarbamate-induced neuronal cell death is mediated by Akt, casein kinase 2, c-Jun N-terminal kinase, and I kappa B kinase in embryonic hippocampal progenitor cells
    • Min YK, Park JH, Chang SA, Kim YS, Alm -YS, Seo JT, et al. Pyrrolidine dithiocarbamate-induced neuronal cell death is mediated by Akt, casein kinase 2, c-Jun N-terminal kinase, and I kappa B kinase in embryonic hippocampal progenitor cells. J Neurosci Res 2003; 71: 689-700.
    • (2003) J Neurosci Res , vol.71 , pp. 689-700
    • Min, Y.K.1    Park, J.H.2    Chang, S.A.3    Kim, Y.S.4    Alm, Y.S.5    Seo, J.T.6
  • 141
    • 16644386249 scopus 로고    scopus 로고
    • Diethyldithiocarbamate can induce two different type of death: Apoptosis and necrosis mediating the differential MAP kinase activation and redox regulation in HL60 cells
    • Kimoto-Kinoshita S, Nishida S, Tomura TT. Diethyldithiocarbamate can induce two different type of death: Apoptosis and necrosis mediating the differential MAP kinase activation and redox regulation in HL60 cells. Mol Cell Biochem 2004; 265: 123 -32.
    • (2004) Mol Cell Biochem , vol.265 , pp. 123-132
    • Kimoto-Kinoshita, S.1    Nishida, S.2    Tomura, T.T.3
  • 142
    • 0242718916 scopus 로고    scopus 로고
    • Novel biphasic effect of pyrrolidine dithiocarbamate on neuronal cell viability is mediated by the differential regulation of intracellular zinc and copper ion levels, NF-κB, and MAP kinases
    • Chung KC, Park JH, Kim CH, Lee HW, Sato N, Uchiyama Y, et al. Novel biphasic effect of pyrrolidine dithiocarbamate on neuronal cell viability is mediated by the differential regulation of intracellular zinc and copper ion levels, NF-κB, and MAP kinases. J Neurosci Res 2000; 59: 117-25.
    • (2000) J Neurosci Res , vol.59 , pp. 117-125
    • Chung, K.C.1    Park, J.H.2    Kim, C.H.3    Lee, H.W.4    Sato, N.5    Uchiyama, Y.6
  • 144
    • 33646030654 scopus 로고    scopus 로고
    • The superoxide dismutase inhibitor diethyldithiocarbamate has antagonistic effects on apoptosis by triggering both cytochrome c release and caspase inhibition
    • Dumay A, Rincheval V, Trotot P, Mignotte B, Vayssiere JL. The superoxide dismutase inhibitor diethyldithiocarbamate has antagonistic effects on apoptosis by triggering both cytochrome c release and caspase inhibition. Free Radic Biol Mod 2006; 40: 1377-90.
    • (2006) Free Radic Biol Mod , vol.40 , pp. 1377-1390
    • Dumay, A.1    Rincheval, V.2    Trotot, P.3    Mignotte, B.4    Vayssiere, J.L.5
  • 145
    • 0942268191 scopus 로고    scopus 로고
    • Does disulfiram have a role in alcoholism treatment today?
    • Fuller RK, Gordis E. Does disulfiram have a role in alcoholism treatment today? Addiction 2004; 99: 21-4
    • (2004) Addiction , vol.99 , pp. 21-24
    • Fuller, R.K.1    Gordis, E.2
  • 146
    • 0032897832 scopus 로고    scopus 로고
    • Safety issues concerning the use of disuffiram in treating alcohol dependence
    • Chick J. Safety issues concerning the use of disuffiram in treating alcohol dependence. Drug Saf 1999; 20: 427-35.
    • (1999) Drug Saf , vol.20 , pp. 427-435
    • Chick, J.1
  • 147
    • 0026755976 scopus 로고
    • Drug-induced hepatic injury - an analysis of 1100 cases reported to the Danish Committee on Adverse Drug -Reactions between 1978 and 1987
    • Friis H, Andreasen PB. Drug-induced hepatic injury - an analysis of 1100 cases reported to the Danish Committee on Adverse Drug -Reactions between 1978 and 1987. J Intern Med 1992; 232: 133-8.
    • (1992) J Intern Med , vol.232 , pp. 133-138
    • Friis, H.1    Andreasen, P.B.2
  • 148
    • 0023798661 scopus 로고
    • Disulfiram-induced fulminating hepatitis - guidelines for liver panel monitoring
    • Wright C, Vafier JA, Lake CR. Disulfiram-induced fulminating hepatitis - guidelines for liver panel monitoring. J Clin Psychiatry 1988; 49: 430-4.
    • (1988) J Clin Psychiatry , vol.49 , pp. 430-434
    • Wright, C.1    Vafier, J.A.2    Lake, C.R.3
  • 150
    • 0034616637 scopus 로고    scopus 로고
    • Blockage of drug resistance in vitro by disulfiram, a drug used to treat alcoholism
    • Loo TW, Clarke DM. Blockage of drug resistance in vitro by disulfiram, a drug used to treat alcoholism. J Natl Cancer Inst 2000; 92: 898-902
    • (2000) J Natl Cancer Inst , vol.92 , pp. 898-902
    • Loo, T.W.1    Clarke, D.M.2
  • 151
    • 1342308328 scopus 로고    scopus 로고
    • The molecular basis of the action of disuffiram as a modulator of the multidrug resistance-linked ATP binding cassette transporters MDR1 (ABCB1) and MRP1 (ABCCI)
    • Sauna ZE, Peng XCH, Nandigarna K, Tekle S, Ambudkar SV. The molecular basis of the action of disuffiram as a modulator of the multidrug resistance-linked ATP binding cassette transporters MDR1 (ABCB1) and MRP1 (ABCCI). Mol Pharmacol 2004; 65: 675-84.
    • (2004) Mol Pharmacol , vol.65 , pp. 675-684
    • Sauna, Z.E.1    Peng, X.C.H.2    Nandigarna, K.3    Tekle, S.4    Ambudkar, S.V.5
  • 152
    • 6044265703 scopus 로고    scopus 로고
    • Disuffiram is a potent modulator of multidrug transporter Cdr1p of Candida albicans
    • Shukla S, Sauna ZE, Prasad R, Ambudkar SV. Disuffiram is a potent modulator of multidrug transporter Cdr1p of Candida albicans. Biochem Biophys Commun 2004; 322: 520-5.
    • (2004) Biochem Biophys Commun , vol.322 , pp. 520-525
    • Shukla, S.1    Sauna, Z.E.2    Prasad, R.3    Ambudkar, S.V.4
  • 153
    • 33644843388 scopus 로고    scopus 로고
    • Disulfiram, an old drug with new potential therapeutic uses for human cancers, and fungal infections
    • Sauna ZE, Shukla S, Ambudkar SV. Disulfiram, an old drug with new potential therapeutic uses for human cancers, and fungal infections. Mol Biosyst 2005; 1: 127-34.
    • (2005) Mol Biosyst , vol.1 , pp. 127-134
    • Sauna, Z.E.1    Shukla, S.2    Ambudkar, S.V.3
  • 154
    • 0029874138 scopus 로고    scopus 로고
    • The NF-κB and IκB proteins: New discoveries and insights
    • Baldwin AS. The NF-κB and IκB proteins: New discoveries and insights. Annu Rev Immunol 1996; 14: 649-83.
    • (1996) Annu Rev Immunol , vol.14 , pp. 649-683
    • Baldwin, A.S.1
  • 157
    • 0037496130 scopus 로고    scopus 로고
    • Transducing signals from antigen receptors to nuclear factor κB
    • Ruland J, Mak TW. Transducing signals from antigen receptors to nuclear factor κB. Immunol Rev 2003; 193: 93-100.
    • (2003) Immunol Rev , vol.193 , pp. 93-100
    • Ruland, J.1    Mak, T.W.2
  • 158
    • 0038605236 scopus 로고    scopus 로고
    • New EMBO member's review - NF-κB and virus infection: Who controls whom
    • Santoro MG, Rossi A, Amici C. New EMBO member's review - NF-κB and virus infection: Who controls whom. EMBO J 2003; 22: 2552-60.
    • (2003) EMBO J , vol.22 , pp. 2552-2560
    • Santoro, M.G.1    Rossi, A.2    Amici, C.3
  • 159
    • 1642553460 scopus 로고    scopus 로고
    • To be, or not to be: NF-κB is the answer: Role of Rel/NF-κB in the regulation of apoptosis
    • Kucharczak J, Simmons MJ, Fan YJ, Gelinas C. To be, or not to be: NF-κB is the answer: Role of Rel/NF-κB in the regulation of apoptosis. Oncogene 2003; 22: 8961-92.
    • (2003) Oncogene , vol.22 , pp. 8961-8992
    • Kucharczak, J.1    Simmons, M.J.2    Fan, Y.J.3    Gelinas, C.4
  • 160
    • 0344845002 scopus 로고    scopus 로고
    • Dying for NF-κB? Control of cell death by transcriptional regulation of the apoptotic machinery
    • Burstein E, Ducrett CS. Dying for NF-κB? Control of cell death by transcriptional regulation of the apoptotic machinery. Curr Open Cell Biol 2003; 15: 732-1.
    • (2003) Curr Open Cell Biol , vol.15 , pp. 732-731
    • Burstein, E.1    Ducrett, C.S.2
  • 162
    • 2342464085 scopus 로고    scopus 로고
    • The two NF-κB activation pathways and their role in innate and adaptive immunity
    • Bonizzi G, Karin M. The two NF-κB activation pathways and their role in innate and adaptive immunity. Trends Immunol 2004; 25: 280-8
    • (2004) Trends Immunol , vol.25 , pp. 280-288
    • Bonizzi, G.1    Karin, M.2
  • 163
    • 3843052483 scopus 로고    scopus 로고
    • Kumar A, Takada Y, Boriek AM, Aggarwal BB. Nuclear factor-κB: Its role in health and disease. J Mol Mod 2004; 82: 43448.
    • Kumar A, Takada Y, Boriek AM, Aggarwal BB. Nuclear factor-κB: Its role in health and disease. J Mol Mod 2004; 82: 43448.
  • 164
    • 11144249792 scopus 로고    scopus 로고
    • Physiological functions for brain NF-κB
    • Meffert MK, Baltimore D. Physiological functions for brain NF-κB. Trends Neurosci 2005; 28: 37-43.
    • (2005) Trends Neurosci , vol.28 , pp. 37-43
    • Meffert, M.K.1    Baltimore, D.2
  • 165
    • 18244379306 scopus 로고    scopus 로고
    • Interactions of NF-κB with chromatin: The art of being at the right place at the right time
    • Natoli G, Saccani S, Bosisio D, Marazzi I. Interactions of NF-κB with chromatin: The art of being at the right place at the right time. Nat Immunol 2005; 6: 439-45.
    • (2005) Nat Immunol , vol.6 , pp. 439-445
    • Natoli, G.1    Saccani, S.2    Bosisio, D.3    Marazzi, I.4
  • 166
    • 20644455960 scopus 로고    scopus 로고
    • Control of lymphocyte development by NF-κB
    • Siebenlist U, Brown K, Claudio E. Control of lymphocyte development by NF-κB. Nat Rev Immunol 2005; 5: 435-45.
    • (2005) Nat Rev Immunol , vol.5 , pp. 435-445
    • Siebenlist, U.1    Brown, K.2    Claudio, E.3
  • 167
    • 23844485308 scopus 로고    scopus 로고
    • The NF-κB signaling pathway in human genetic diseases
    • Courtois G. The NF-κB signaling pathway in human genetic diseases. Cell Mol Life Sci 2005; 62: 1682-91.
    • (2005) Cell Mol Life Sci , vol.62 , pp. 1682-1691
    • Courtois, G.1
  • 168
    • 28544453229 scopus 로고    scopus 로고
    • Role of NF-κB in the immune system and bone
    • Jimi E, Ghoso S. Role of NF-κB in the immune system and bone. Immumol Rev 2005; 208: 80-7.
    • (2005) Immumol Rev , vol.208 , pp. 80-87
    • Jimi, E.1    Ghoso, S.2
  • 169
    • 33745852665 scopus 로고    scopus 로고
    • Alternative pathways of NF-κB activation: A double-edged sword in health and disease
    • Xiao GT, Rabson AB, Young W, Qing GL, Qu ZX. Alternative pathways of NF-κB activation: A double-edged sword in health and disease. Cytokine Growth Factor Rev 2006; 17: 281-93.
    • (2006) Cytokine Growth Factor Rev , vol.17 , pp. 281-293
    • Xiao, G.T.1    Rabson, A.B.2    Young, W.3    Qing, G.L.4    Qu, Z.X.5
  • 170
    • 33749055328 scopus 로고    scopus 로고
    • Memet S. NF-κB functions in the nervous system: From development to disease. Biochem Pharmacol 2006; 72: 1180-95.
    • Memet S. NF-κB functions in the nervous system: From development to disease. Biochem Pharmacol 2006; 72: 1180-95.
  • 171
    • 33749265867 scopus 로고    scopus 로고
    • The alternative NF-κB pathway from biochemistry to biology: Pitfalls and promises for future drug development
    • Dejardin E. The alternative NF-κB pathway from biochemistry to biology: Pitfalls and promises for future drug development. Biochem Pharmacol 2006; 72: 1161-79.
    • (2006) Biochem Pharmacol , vol.72 , pp. 1161-1179
    • Dejardin, E.1
  • 172
    • 33750466230 scopus 로고    scopus 로고
    • Introduction to NF-βB: Players, pathways, perspectives
    • Gilmore TD. Introduction to NF-βB: Players, pathways, perspectives. Oncogene 2006; 25: 6680-4.
    • (2006) Oncogene , vol.25 , pp. 6680-6684
    • Gilmore, T.D.1
  • 173
    • 33750448661 scopus 로고    scopus 로고
    • Transcriptional regulation via the, NF-κB signaling module
    • Hoffmann A, Natoli G, Ghosh G. Transcriptional regulation via the, NF-κB signaling module. Oncogene 2006; 6706-16.
    • (2006) Oncogene , pp. 6706-6716
    • Hoffmann, A.1    Natoli, G.2    Ghosh, G.3
  • 174
    • 33750435582 scopus 로고    scopus 로고
    • NF-βB and the immune response
    • Hayden MS, West AP, Ghosh S. NF-βB and the immune response. Oncogene 2006; 25: 6758-80.
    • (2006) Oncogene , vol.25 , pp. 6758-6780
    • Hayden, M.S.1    West, A.P.2    Ghosh, S.3
  • 175
    • 33750475618 scopus 로고    scopus 로고
    • Current insights into the regulation of programmed cell death by NF-κB
    • Dutta J, Fan X, Gupta N, Fan G, Gelinas C. Current insights into the regulation of programmed cell death by NF-κB. Oncogene 2006; 25: 6800-16.
    • (2006) Oncogene , vol.25 , pp. 6800-6816
    • Dutta, J.1    Fan, X.2    Gupta, N.3    Fan, G.4    Gelinas, C.5
  • 176
    • 33750475959 scopus 로고    scopus 로고
    • Cross-talk between nuclear receptors and NF-κB
    • De Basscher K, Vanden Berghe W, Haegeman G. Cross-talk between nuclear receptors and NF-κB. Oncogene 2006; 25: 6868-86.
    • (2006) Oncogene , vol.25 , pp. 6868-6886
    • De Basscher, K.1    Vanden Berghe, W.2    Haegeman, G.3
  • 177
    • 0030615201 scopus 로고    scopus 로고
    • Mechanisms of disease. Nuclear factor-κB - a pívótal transcription factor in chronic inflammatory disease
    • Barnes PJ, Larin M. Mechanisms of disease. Nuclear factor-κB - a pívótal transcription factor in chronic inflammatory disease. N Engl J Med 1997; 336: 1066-71.
    • (1997) N Engl J Med , vol.336 , pp. 1066-1071
    • Barnes, P.J.1    Larin, M.2
  • 178
    • 17544365995 scopus 로고    scopus 로고
    • The IκB/NF-κB system: A key determinant of mucosal inflammation and protection
    • Jobin C, Sartor RB. The IκB/NF-κB system: A key determinant of mucosal inflammation and protection. Am J Physiol Cell Physiol 2000; 278: C451-62.
    • (2000) Am J Physiol Cell Physiol , vol.278
    • Jobin, C.1    Sartor, R.B.2
  • 179
    • 0033623536 scopus 로고    scopus 로고
    • Makarov SS. NF-κB as a therapeutic target in chronic inflammation: recent advances. Mol Med Today 2000; 6: 441-8.
    • Makarov SS. NF-κB as a therapeutic target in chronic inflammation: recent advances. Mol Med Today 2000; 6: 441-8.
  • 180
    • 0034942860 scopus 로고    scopus 로고
    • Makarov SS. NF-κB in rheumatoid arthritis: A pivotal regulator of inflammation, hyperplasia, and tissue destruction. Arthritis Res 2001; 3: 200-6.
    • Makarov SS. NF-κB in rheumatoid arthritis: A pivotal regulator of inflammation, hyperplasia, and tissue destruction. Arthritis Res 2001; 3: 200-6.
  • 181
    • 0346363234 scopus 로고    scopus 로고
    • Inflammation meets oxidation: NF-κB as a mediator of initial lesion development in atherosclerosis
    • Kutuk O, Basaga H. Inflammation meets oxidation: NF-κB as a mediator of initial lesion development in atherosclerosis. Trends Mol Med 2003; 9: 549-57.
    • (2003) Trends Mol Med , vol.9 , pp. 549-557
    • Kutuk, O.1    Basaga, H.2
  • 182
    • 1342345219 scopus 로고    scopus 로고
    • Monaco C, Paleolog E. Nuclear factor κB: A potential therapeutic target in atherosclerosis and thrombosis. Cardiovasc Res 2004; 61: 671-82.
    • Monaco C, Paleolog E. Nuclear factor κB: A potential therapeutic target in atherosclerosis and thrombosis. Cardiovasc Res 2004; 61: 671-82.
  • 183
    • 4444275699 scopus 로고    scopus 로고
    • The NF-κB pathway as a potential target for autoimmune disease therapy
    • Bacher S, Schmitz ML. The NF-κB pathway as a potential target for autoimmune disease therapy. Curr Pharm Des 2004; 10: 2827-37.
    • (2004) Curr Pharm Des , vol.10 , pp. 2827-2837
    • Bacher, S.1    Schmitz, M.L.2
  • 186
    • 33645116785 scopus 로고    scopus 로고
    • NF-κB activation as a pathological mechanism of septic shock and inflammation
    • Liu SF, Malik AB. NF-κB activation as a pathological mechanism of septic shock and inflammation. Am J Physiol Lung Cell Mol Physiol 2006; 290: L622-45.
    • (2006) Am J Physiol Lung Cell Mol Physiol , vol.290
    • Liu, S.F.1    Malik, A.B.2
  • 187
    • 33744825413 scopus 로고    scopus 로고
    • Nuclear factor kappa B is a promising therapeutic target in inflammatory lung disease
    • Park GY, Christman JW. Nuclear factor kappa B is a promising therapeutic target in inflammatory lung disease. Curr Drug Targets 2006; 7: 661-8
    • (2006) Curr Drug Targets , vol.7 , pp. 661-668
    • Park, G.Y.1    Christman, J.W.2
  • 188
    • 33746480903 scopus 로고    scopus 로고
    • NF-κB as a potential therapeutic target in osteoarthritis and rheumatoid arthritis
    • Roman-Blas JA, Jimenez SA. NF-κB as a potential therapeutic target in osteoarthritis and rheumatoid arthritis. Osteoarthritis Cartilage 2006; 14: 839-48.
    • (2006) Osteoarthritis Cartilage , vol.14 , pp. 839-848
    • Roman-Blas, J.A.1    Jimenez, S.A.2
  • 189
    • 33646549937 scopus 로고    scopus 로고
    • Tuning up inflammation: How DNA sequence and chromatin organization control the induction of inflammatory genes by NF-κB
    • Natoli G. Tuning up inflammation: How DNA sequence and chromatin organization control the induction of inflammatory genes by NF-κB. FEBS Lett 2006; 580: 2843-9.
    • (2006) FEBS Lett , vol.580 , pp. 2843-2849
    • Natoli, G.1
  • 191
    • 0029671333 scopus 로고    scopus 로고
    • NF-κB-mediated chromatin reconfiguration and transcriptional activation of the HIV-1 enhancer in vitro
    • Pazin MJ, Sheridan PL, Cannon K, Cao ZD, Keck JG, Kadonaga JT, et al. NF-κB-mediated chromatin reconfiguration and transcriptional activation of the HIV-1 enhancer in vitro. Genes Dev 1996; 10: 37-49.
    • (1996) Genes Dev , vol.10 , pp. 37-49
    • Pazin, M.J.1    Sheridan, P.L.2    Cannon, K.3    Cao, Z.D.4    Keck, J.G.5    Kadonaga, J.T.6
  • 193
    • 0034892960 scopus 로고    scopus 로고
    • Activation of human immunodeficiency virus transcription in T cells revisited: NF-κB p65 stimulates transcriptional elongation
    • West MJ, Lowe AD, Kam J. Activation of human immunodeficiency virus transcription in T cells revisited: NF-κB p65 stimulates transcriptional elongation. J Virol 2001; 75: 8524-37.
    • (2001) J Virol , vol.75 , pp. 8524-8537
    • West, M.J.1    Lowe, A.D.2    Kam, J.3
  • 194
    • 0041308422 scopus 로고    scopus 로고
    • Pande V, Ramos MJ. Nuclear factor κB: A potential target for anti-HIV chemotherapy. Curr Med Chem 2003; 10: 1603-15.
    • Pande V, Ramos MJ. Nuclear factor κB: A potential target for anti-HIV chemotherapy. Curr Med Chem 2003; 10: 1603-15.
  • 195
    • 30444431914 scopus 로고    scopus 로고
    • NF-κB p50 promotes HIV latency through HDAC recruitment and repression of transcriptional initiation
    • Williams SA, Chen LF, Kwon H, Ruiz-Jarabo CM, Verdin E, Greene WC. NF-κB p50 promotes HIV latency through HDAC recruitment and repression of transcriptional initiation. EMIBO J 2006; 25: 139-49.
    • (2006) EMIBO J , vol.25 , pp. 139-149
    • Williams, S.A.1    Chen, L.F.2    Kwon, H.3    Ruiz-Jarabo, C.M.4    Verdin, E.5    Greene, W.C.6
  • 197
    • 0037096221 scopus 로고    scopus 로고
    • Arresting a major culprit in cancer
    • Haefner B. NF-κB: Arresting a major culprit in cancer. Drug Discov Today 2002; 7: 653-63.
    • (2002) Drug Discov Today , vol.7 , pp. 653-663
    • Haefner, B.1
  • 198
    • 85047699198 scopus 로고    scopus 로고
    • Nuclear transcription factor-κB as a target for cancer drug development
    • Garg A, Aggarwal BB. Nuclear transcription factor-κB as a target for cancer drug development. Leukemia 2002; 16: 1053-68.
    • (2002) Leukemia , vol.16 , pp. 1053-1068
    • Garg, A.1    Aggarwal, B.B.2
  • 199
    • 0036717195 scopus 로고    scopus 로고
    • chemokine gene transcription and tumour growth
    • Richmond A. NF-κB, chemokine gene transcription and tumour growth. Nat Rev Immunol 2002; 2: 664-74.
    • (2002) Nat Rev Immunol , vol.2 , pp. 664-674
    • Richmond, A.1
  • 200
    • 0037328613 scopus 로고    scopus 로고
    • NF-κB activation in cancer: A challenge for ubiquitination- and proteasome-based therapeutic approach
    • Amit S, Ben-Neriah Y. NF-κB activation in cancer: A challenge for ubiquitination- and proteasome-based therapeutic approach. Semin Cancer Biol 2003; 13: 15-28.
    • (2003) Semin Cancer Biol , vol.13 , pp. 15-28
    • Amit, S.1    Ben-Neriah, Y.2
  • 201
    • 0038724819 scopus 로고    scopus 로고
    • Deregulation of NF-κB and its upstream kinases in cancer
    • Sun SC, Xiao GT. Deregulation of NF-κB and its upstream kinases in cancer. Cancer Metastasis Rev 2003; 22: 405-22.
    • (2003) Cancer Metastasis Rev , vol.22 , pp. 405-422
    • Sun, S.C.1    Xiao, G.T.2
  • 202
    • 4544342570 scopus 로고    scopus 로고
    • Aggarwal BB. Nuclear factor-κB: The enemy within. Cancer Cell 2004; 6: 203-8.
    • Aggarwal BB. Nuclear factor-κB: The enemy within. Cancer Cell 2004; 6: 203-8.
  • 203
    • 8544283079 scopus 로고    scopus 로고
    • Endogenous inhibitors of nuclear factor-κB, an opportunity for cancer control
    • Chen F. Endogenous inhibitors of nuclear factor-κB, an opportunity for cancer control. Cancer Res 2004; 64: 8135-8.
    • (2004) Cancer Res , vol.64 , pp. 8135-8138
    • Chen, F.1
  • 204
    • 16844366650 scopus 로고    scopus 로고
    • Nuclear factor-κB inhibitors as sensitizers to anticancer drugs
    • Nakanishi C, Toi M. Nuclear factor-κB inhibitors as sensitizers to anticancer drugs. Nat Rev Cancer 2005; 5: 297-309.
    • (2005) Nat Rev Cancer , vol.5 , pp. 297-309
    • Nakanishi, C.1    Toi, M.2
  • 205
    • 25844459154 scopus 로고    scopus 로고
    • Karin M, Greten FR. NF-κB: Linking inflammation and immunity to cancer development and progression. Nat Rev Immunol 2005; 5: 749-59
    • Karin M, Greten FR. NF-κB: Linking inflammation and immunity to cancer development and progression. Nat Rev Immunol 2005; 5: 749-59
  • 206
    • 26444605834 scopus 로고    scopus 로고
    • IKK/NF-κB signaling: Balancing life and death - a new approach to cancer therapy
    • Luo JL, Kamata H, Karin M. IKK/NF-κB signaling: Balancing life and death - a new approach to cancer therapy. J Clip Invest 2005; 115: 2625-32.
    • (2005) J Clip Invest , vol.115 , pp. 2625-2632
    • Luo, J.L.1    Kamata, H.2    Karin, M.3
  • 208
    • 33745298519 scopus 로고    scopus 로고
    • Nuclear factor-κB in cancer development and progression
    • Karin M. Nuclear factor-κB in cancer development and progression. Nature 2006; 441: 431-6.
    • (2006) Nature , vol.441 , pp. 431-436
    • Karin, M.1
  • 209
    • 33750432183 scopus 로고    scopus 로고
    • Nuclear factor-κB and inhibitor of κB kinase pathways in oncogenic initiation and progression
    • Basseres DS, Baldwin AS. Nuclear factor-κB and inhibitor of κB kinase pathways in oncogenic initiation and progression. Oncogene 2006; 25. 6817-30.
    • (2006) Oncogene , vol.25 , pp. 6817-6830
    • Basseres, D.S.1    Baldwin, A.S.2
  • 210
    • 0033979815 scopus 로고    scopus 로고
    • Roles of nuclear factor κB in neuronal survival and plasticity
    • Mattson MP, Culmsee C, Yu ZF, Camandola S. Roles of nuclear factor κB in neuronal survival and plasticity. J Neurochem 2000; 74; 443-56.
    • (2000) J Neurochem , vol.74 , pp. 443-456
    • Mattson, M.P.1    Culmsee, C.2    Yu, Z.F.3    Camandola, S.4
  • 211
    • 0035112978 scopus 로고    scopus 로고
    • NF-κB is involved in the survival of cerebellar granule neurons: Association of IκB phosphorylation with cell survival
    • Koulich E, Nguyen T, Johnson K, Giardina CA, D. Mello SR. NF-κB is involved in the survival of cerebellar granule neurons: Association of IκB phosphorylation with cell survival. J Neurochem 2001; 76: 1188-98.
    • (2001) J Neurochem , vol.76 , pp. 1188-1198
    • Koulich, E.1    Nguyen, T.2    Johnson, K.3    Giardina, C.D.4    Mello, S.R.5
  • 212
    • 0035399856 scopus 로고    scopus 로고
    • Activation of the nuclear factor-κB is a key event in brain tolerance
    • Blondeau N, Widmann C, Lazdunski M, Heurteauk C. Activation of the nuclear factor-κB is a key event in brain tolerance. J Neurosci 2001; 21: 4668-77.
    • (2001) J Neurosci , vol.21 , pp. 4668-4677
    • Blondeau, N.1    Widmann, C.2    Lazdunski, M.3    Heurteauk, C.4
  • 213
    • 0035892721 scopus 로고    scopus 로고
    • Inhibition of nuclear factor-κB activation induces apoptosis in cerebellar granule cells
    • Piccioli P, Porcile C, Stanzione S, Bisaglia M, Bajetto A, Bonavia R, et al. Inhibition of nuclear factor-κB activation induces apoptosis in cerebellar granule cells. J Neurosci Res 2001; 66: 1064-73.
    • (2001) J Neurosci Res , vol.66 , pp. 1064-1073
    • Piccioli, P.1    Porcile, C.2    Stanzione, S.3    Bisaglia, M.4    Bajetto, A.5    Bonavia, R.6
  • 214
    • 0037266092 scopus 로고    scopus 로고
    • Yalcin A, Koulich E, Mohamed S, Liu L, D'Mello SR. Apoptosis in cerebellar granule neurons is associated with reduced interaction between CREB-binding protein and NF-κB. J Neurochem 2003; 84: 397-408.
    • Yalcin A, Koulich E, Mohamed S, Liu L, D'Mello SR. Apoptosis in cerebellar granule neurons is associated with reduced interaction between CREB-binding protein and NF-κB. J Neurochem 2003; 84: 397-408.
  • 215
    • 0037317077 scopus 로고    scopus 로고
    • Activation of the transcription factor NF-κB in Schwann cells is required for peripheral myelin formation
    • Nickols JC, Valentine W, Kanwal S, Carter BD. Activation of the transcription factor NF-κB in Schwann cells is required for peripheral myelin formation. Nat Neurosci 2003; 6: 161-7.
    • (2003) Nat Neurosci , vol.6 , pp. 161-167
    • Nickols, J.C.1    Valentine, W.2    Kanwal, S.3    Carter, B.D.4
  • 216
    • 17744371866 scopus 로고    scopus 로고
    • NF-kappa B signalling regulates the growth of neural processes in the developing PNS and CNS
    • Gutierrez H, Hale VA, Dolcet X, Davies A. NF-kappa B signalling regulates the growth of neural processes in the developing PNS and CNS. Development 2005; 132: 1713-26.
    • (2005) Development , vol.132 , pp. 1713-1726
    • Gutierrez, H.1    Hale, V.A.2    Dolcet, X.3    Davies, A.4
  • 217
    • 33845317629 scopus 로고    scopus 로고
    • Action of NF-κB on the delta opioid receptor gene promoter
    • Chen YLL, Law PY, Loh HH. Action of NF-κB on the delta opioid receptor gene promoter. Biochem Biophys Res Commun 2007; 352: 818-22
    • (2007) Biochem Biophys Res Commun , vol.352 , pp. 818-822
    • Chen, Y.L.L.1    Law, P.Y.2    Loh, H.H.3
  • 218
    • 4444279747 scopus 로고    scopus 로고
    • Of mice and men. Meeting on the biology and pathology of NF-κB
    • Ben-Neriah Y, Schmitz ML. Of mice and men. Meeting on the biology and pathology of NF-κB. EMBO Rep 2004; 5: 668-73.
    • (2004) EMBO Rep , vol.5 , pp. 668-673
    • Ben-Neriah, Y.1    Schmitz, M.L.2
  • 219
    • 4544228457 scopus 로고    scopus 로고
    • Functions of NF-κB1 and NF-κB2 in immune cell biology
    • Beinke S, Ley SC. Functions of NF-κB1 and NF-κB2 in immune cell biology. Biochem J 2004; 382: 393-409.
    • (2004) Biochem J , vol.382 , pp. 393-409
    • Beinke, S.1    Ley, S.C.2
  • 222
    • 0031126702 scopus 로고    scopus 로고
    • Rel/NF-κB and IκB proteins: An overview
    • May MJ, Ghosh S. Rel/NF-κB and IκB proteins: An overview. Semin Cancer Biol 1997; 8: 63-73.
    • (1997) Semin Cancer Biol , vol.8 , pp. 63-73
    • May, M.J.1    Ghosh, S.2
  • 223
    • 0032217267 scopus 로고    scopus 로고
    • Baeuerle PA. IκB-NF-κB structures: At the interface of inflammation control. Cell 1998; 95: 729-31.
    • Baeuerle PA. IκB-NF-κB structures: At the interface of inflammation control. Cell 1998; 95: 729-31.
  • 224
    • 0031906550 scopus 로고    scopus 로고
    • Signal transduction through NF-κB
    • May MJ, Ghosh S. Signal transduction through NF-κB. Immunol Today 1998; 19: 80-8.
    • (1998) Immunol Today , vol.19 , pp. 80-88
    • May, M.J.1    Ghosh, S.2
  • 226
    • 33747065618 scopus 로고    scopus 로고
    • Israel A. NF-κB activation: Nondegradative ubiquitination implicates NEMO, Trends Immunol 2006; 27: 395-7.
    • Israel A. NF-κB activation: Nondegradative ubiquitination implicates NEMO, Trends Immunol 2006; 27: 395-7.
  • 227
    • 33750312876 scopus 로고    scopus 로고
    • Posttranslational modifications of NEMO and its partners in NF-κB signaling
    • Sebban H, Yamaoka S, Courtois G. Posttranslational modifications of NEMO and its partners in NF-κB signaling. Trends Cell Biol 2006; 16: 569-77.
    • (2006) Trends Cell Biol , vol.16 , pp. 569-577
    • Sebban, H.1    Yamaoka, S.2    Courtois, G.3
  • 228
    • 33750443289 scopus 로고    scopus 로고
    • IκB kinase complexes: Gateways to NF-κB activation and transcription
    • Scheidereit C. IκB kinase complexes: Gateways to NF-κB activation and transcription. Oncogene 2006; 25: 6685-705.
    • (2006) Oncogene , vol.25 , pp. 6685-6705
    • Scheidereit, C.1
  • 229
    • 33845768987 scopus 로고    scopus 로고
    • Integrating cell-signalling pathways with NF-κB and IKK function
    • Perkins ND. Integrating cell-signalling pathways with NF-κB and IKK function. Nat Rev Mol Cell Biol 2007; 8: 49-62.
    • (2007) Nat Rev Mol Cell Biol , vol.8 , pp. 49-62
    • Perkins, N.D.1
  • 230
    • 0033106149 scopus 로고    scopus 로고
    • A ubiquitin ligase complex essential for the NF-κB, Wnt/Wingless; and Hedgehog signaling pathways
    • Maniatis T. A ubiquitin ligase complex essential for the NF-κB, Wnt/Wingless; and Hedgehog signaling pathways. Genes Dev 1999; 13: 505-10.
    • (1999) Genes Dev , vol.13 , pp. 505-510
    • Maniatis, T.1
  • 231
    • 0036234459 scopus 로고    scopus 로고
    • Missing pieces in NF-κB puzzle
    • Ghosh S, Karin M. Missing pieces in NF-κB puzzle Cell 2002; 109: S81-96.
    • (2002) Cell , vol.109
    • Ghosh, S.1    Karin, M.2
  • 232
    • 4444376712 scopus 로고    scopus 로고
    • Signaling to NF-κB
    • Hayden MS, Ghosh S. Signaling to NF-κB. Genes Dev 2004; 18: 2195-224.
    • (2004) Genes Dev , vol.18 , pp. 2195-2224
    • Hayden, M.S.1    Ghosh, S.2
  • 233
    • 2342522110 scopus 로고    scopus 로고
    • Shaping the nuclear action of NF-κB
    • Chen LF, Greene WC. Shaping the nuclear action of NF-κB. Nat Rev Mol Cell Biol 2004; 5: 392-401.
    • (2004) Nat Rev Mol Cell Biol , vol.5 , pp. 392-401
    • Chen, L.F.1    Greene, W.C.2
  • 234
    • 0026590541 scopus 로고
    • Dithiocarbamates as potent inhibitors of nuclear factor κB activation in intact cells
    • Schreck R, Meier B, Mannel DN, Droge W, Baeuerle PA. Dithiocarbamates as potent inhibitors of nuclear factor κB activation in intact cells. J Exp Med 1992; 175: 1181-94.
    • (1992) J Exp Med , vol.175 , pp. 1181-1194
    • Schreck, R.1    Meier, B.2    Mannel, D.N.3    Droge, W.4    Baeuerle, P.A.5
  • 235
    • 0027176524 scopus 로고
    • Rapid proteolysis of IκB-α is necessary for activation of transcription factor NF-κB
    • Henkel T, Machleidt T, Alkalay I, Kronke M, Ben-Neriah Y, Baeuerle PA. Rapid proteolysis of IκB-α is necessary for activation of transcription factor NF-κB. Nature 1993; 365: 182-5.
    • (1993) Nature , vol.365 , pp. 182-185
    • Henkel, T.1    Machleidt, T.2    Alkalay, I.3    Kronke, M.4    Ben-Neriah, Y.5    Baeuerle, P.A.6
  • 236
    • 0001047586 scopus 로고    scopus 로고
    • Pyrrolidine dithiocarbamate prevents I-κB degradation and reduces microvascular injury induced by lipopolysaccharide in multiple organs
    • Liu SF, Ye XB, Malik AB. Pyrrolidine dithiocarbamate prevents I-κB degradation and reduces microvascular injury induced by lipopolysaccharide in multiple organs. Mol Pharmacol 1999; 55: 658-67
    • (1999) Mol Pharmacol , vol.55 , pp. 658-667
    • Liu, S.F.1    Ye, X.B.2    Malik, A.B.3
  • 237
    • 0031572429 scopus 로고    scopus 로고
    • In vivo inhibition of nuclear factor-κB activation prevents inducible nitric oxide synthase expression and systemic hypotension in a rat model of septic shock
    • Liu SF, Ye XB, Malik AB. In vivo inhibition of nuclear factor-κB activation prevents inducible nitric oxide synthase expression and systemic hypotension in a rat model of septic shock. J Immunol 1997; 159:3976-83.
    • (1997) J Immunol , vol.159 , pp. 3976-3983
    • Liu, S.F.1    Ye, X.B.2    Malik, A.B.3
  • 238
    • 0033592288 scopus 로고    scopus 로고
    • Inhibition of NF-κB activation by pyrrolidine dithiocarbamate prevents in vivo expression of proinflammatory genes
    • Liu SF, Ye XB, Malik AB. Inhibition of NF-κB activation by pyrrolidine dithiocarbamate prevents in vivo expression of proinflammatory genes. Circulation 1999; 100: 1330-7.
    • (1999) Circulation , vol.100 , pp. 1330-1337
    • Liu, S.F.1    Ye, X.B.2    Malik, A.B.3
  • 240
    • 0036174993 scopus 로고    scopus 로고
    • Pyrrolidine dithiocarbamate attenuates the development of acute and-chronic inflammation
    • Cuzzocrea S, Chatterjee PK, Mazzon E, Dugo L, Serraino I, Britti D, et al. Pyrrolidine dithiocarbamate attenuates the development of acute and-chronic inflammation. Br J Pharmacol 2002; 135: 496-510.
    • (2002) Br J Pharmacol , vol.135 , pp. 496-510
    • Cuzzocrea, S.1    Chatterjee, P.K.2    Mazzon, E.3    Dugo, L.4    Serraino, I.5    Britti, D.6
  • 241
    • 0036190630 scopus 로고    scopus 로고
    • Inhibitory effects of pyrrolidine dithiocarbamate on endotoxin-induced uveitis in Lewis rats
    • Ohta K, Nakayama K, Kurokawa T, Kikuchi T, Yoshimura N. Inhibitory effects of pyrrolidine dithiocarbamate on endotoxin-induced uveitis in Lewis rats. Invest Ophthalmol Vis Sci 2002; 43: 744-50.
    • (2002) Invest Ophthalmol Vis Sci , vol.43 , pp. 744-750
    • Ohta, K.1    Nakayama, K.2    Kurokawa, T.3    Kikuchi, T.4    Yoshimura, N.5
  • 242
    • 0036186392 scopus 로고    scopus 로고
    • Pyrrolidine dithiocarbamate protects against thioacetamide-induced failure in rats
    • Bruck R, Aeed H, Schey R, Matas Z, Reifen R, Zaiger G, et al. Pyrrolidine dithiocarbamate protects against thioacetamide-induced failure in rats. J Hepatol 2002; 36: 370-7.
    • (2002) J Hepatol , vol.36 , pp. 370-377
    • Bruck, R.1    Aeed, H.2    Schey, R.3    Matas, Z.4    Reifen, R.5    Zaiger, G.6
  • 243
    • 6444245024 scopus 로고    scopus 로고
    • Inhibition of nuclear factor-κB activation by pyrrolidine dithio-carbamate prevents chronic FK506 nephropathy
    • Tamada S, Nakatani T, Asai T, Tashiro K, Komiya T, Sumi T, et al. Inhibition of nuclear factor-κB activation by pyrrolidine dithio-carbamate prevents chronic FK506 nephropathy. Kidney Int 2003; 63: 306-14.
    • (2003) Kidney Int , vol.63 , pp. 306-314
    • Tamada, S.1    Nakatani, T.2    Asai, T.3    Tashiro, K.4    Komiya, T.5    Sumi, T.6
  • 245
    • 10744221054 scopus 로고    scopus 로고
    • Pyrrolidine dithiocarbamate attenuates the development of organ failure induced by zymosan in mice
    • Cuzzocrea S, Rossi A, Pisano B, Di Paola R, Genovese T, Patel NSA, et al. Pyrrolidine dithiocarbamate attenuates the development of organ failure induced by zymosan in mice. Intensive Care Med 2003; 29: 2016-25.
    • (2003) Intensive Care Med , vol.29 , pp. 2016-2025
    • Cuzzocrea, S.1    Rossi, A.2    Pisano, B.3    Di Paola, R.4    Genovese, T.5    Patel, N.S.A.6
  • 246
    • 7944227871 scopus 로고    scopus 로고
    • Inhibition of the nuclear factor-κB activation with pyrrolidine dithiocarbamate attenuating inflammation and oxidative stress after experimental spinal cord trauma in rats
    • La Rosa G, Cardali S, Genovese T, Conti A, Di Paola R, Le Torre D, et al. Inhibition of the nuclear factor-κB activation with pyrrolidine dithiocarbamate attenuating inflammation and oxidative stress after experimental spinal cord trauma in rats. J Neurosurg Spine 2004; 1: 311-21.
    • (2004) J Neurosurg Spine , vol.1 , pp. 311-321
    • La Rosa, G.1    Cardali, S.2    Genovese, T.3    Conti, A.4    Di Paola, R.5    Le Torre, D.6
  • 247
    • 18244370287 scopus 로고    scopus 로고
    • Effects of the NF-κB inhibitor pyrrolidine dithiocarbamate on experimentally induced autoimmune anterior uveitis
    • Yang CH, Fang IM, Lin CP, Yang CM, Chen MS. Effects of the NF-κB inhibitor pyrrolidine dithiocarbamate on experimentally induced autoimmune anterior uveitis. Invest Ophthalmol Vis Sci 2005; 46: 1339-47.
    • (2005) Invest Ophthalmol Vis Sci , vol.46 , pp. 1339-1347
    • Yang, C.H.1    Fang, I.M.2    Lin, C.P.3    Yang, C.M.4    Chen, M.S.5
  • 249
    • 33751258581 scopus 로고    scopus 로고
    • Protective effects of pyrrolidine dithiocarbamate on myocardium apoptosis induced by adriamycin in rats
    • Li HL, Gu HY, Sun BG. Protective effects of pyrrolidine dithiocarbamate on myocardium apoptosis induced by adriamycin in rats. Int J Cardiol 2007; 114: 159-65.
    • (2007) Int J Cardiol , vol.114 , pp. 159-165
    • Li, H.L.1    Gu, H.Y.2    Sun, B.G.3
  • 252
    • 0029959753 scopus 로고    scopus 로고
    • Pyrrolidine dithiocarbamate inhibits the production of interleukin-6, interleukin-8, and granulocyte-macrophage colony-stimulating factor by human endothelial cells in response to inflammatory mediators: Modulation of NF-κB and AP-1 transcription factors activity
    • Munoz C, Pascual-Salcedo D, Castellanos MD, Alfranca A, Aragones J, Vara A. et al. Pyrrolidine dithiocarbamate inhibits the production of interleukin-6, interleukin-8, and granulocyte-macrophage colony-stimulating factor by human endothelial cells in response to inflammatory mediators: Modulation of NF-κB and AP-1 transcription factors activity. Blood 1996; 88: 3482-90.
    • (1996) Blood , vol.88 , pp. 3482-3490
    • Munoz, C.1    Pascual-Salcedo, D.2    Castellanos, M.D.3    Alfranca, A.4    Aragones, J.5    Vara, A.6
  • 253
    • 33845973376 scopus 로고    scopus 로고
    • Pyrrolidine dithiocarbamate inhibits interleukin-6 signaling through impaired STAT3 activation and association with transcriptional coactivators in hepatocytes
    • He HJ, Zhu TN, Xie Y, Fan JS, Kole S, Saxena S, et al. Pyrrolidine dithiocarbamate inhibits interleukin-6 signaling through impaired STAT3 activation and association with transcriptional coactivators in hepatocytes. J Biol Chem 2006; 281: 31369-79.
    • (2006) J Biol Chem , vol.281 , pp. 31369-31379
    • He, H.J.1    Zhu, T.N.2    Xie, Y.3    Fan, J.S.4    Kole, S.5    Saxena, S.6
  • 254
    • 33744996941 scopus 로고    scopus 로고
    • The NF-κB inhibitor pyrrolidine dithiocarbarnate blocks IL-1β induced hyaluronan synthase 1 (HAS1) mRNA transcription, pointing at NF-κB dependence of the gene HASI
    • Kao JJ. The NF-κB inhibitor pyrrolidine dithiocarbarnate blocks IL-1β induced hyaluronan synthase 1 (HAS1) mRNA transcription, pointing at NF-κB dependence of the gene HASI. Exp Gerontol 2006; 41: 641-7.
    • (2006) Exp Gerontol , vol.41 , pp. 641-647
    • Kao, J.J.1
  • 255
    • 0029416976 scopus 로고
    • Constitutive nuclear NF-κB/rel DNA-binding activity of rat thymocytes is increased by stimuli that promote apoptosis, but not inhibited by pyrrolidine dithiocarbamate
    • Slater AFG, Kimland M, Jiang SA, Orrenius S. Constitutive nuclear NF-κB/rel DNA-binding activity of rat thymocytes is increased by stimuli that promote apoptosis, but not inhibited by pyrrolidine dithiocarbamate. Biochem J. 1995; 312: 833-8.
    • (1995) Biochem J , vol.312 , pp. 833-838
    • Slater, A.F.G.1    Kimland, M.2    Jiang, S.A.3    Orrenius, S.4
  • 256
    • 0034737474 scopus 로고    scopus 로고
    • Biphasic effects of dithiocarbamates on the activity of nuclear factor-κB
    • Kim CH, Kim JH, Moon SJ, Hsu CY, Seo JT, Ahn YS. Biphasic effects of dithiocarbamates on the activity of nuclear factor-κB. Eur J Pharmacol 2000; 392: 133-6.
    • (2000) Eur J Pharmacol , vol.392 , pp. 133-136
    • Kim, C.H.1    Kim, J.H.2    Moon, S.J.3    Hsu, C.Y.4    Seo, J.T.5    Ahn, Y.S.6
  • 257
    • 0028902123 scopus 로고
    • Modulltion of transcription factor NF-κB activity by intracellular glutathione levels and by variations of the extracellular cysteine supply
    • Mihm S, Galter D, Droge W. Modulltion of transcription factor NF-κB activity by intracellular glutathione levels and by variations of the extracellular cysteine supply. FASEB J 1995; 9: 246-52.
    • (1995) FASEB J , vol.9 , pp. 246-252
    • Mihm, S.1    Galter, D.2    Droge, W.3
  • 258
    • 0036830469 scopus 로고    scopus 로고
    • Regulation of gene expression by oxygen: NF-κB and HIF-1, two extremes
    • Michiels C, Minet E, Mottet D, Raes M. Regulation of gene expression by oxygen: NF-κB and HIF-1, two extremes. Free Radic Biol Med 2002; 33:1231-42.
    • (2002) Free Radic Biol Med , vol.33 , pp. 1231-1242
    • Michiels, C.1    Minet, E.2    Mottet, D.3    Raes, M.4
  • 259
    • 0036893153 scopus 로고    scopus 로고
    • Science review: Redox and oxygen-sensitive transcription factors in the regulation of oxidant-mediated lung injury: Role for nuclear factor-κB
    • Haddad JJ. Science review: Redox and oxygen-sensitive transcription factors in the regulation of oxidant-mediated lung injury: Role for nuclear factor-κB. Crit Care 2002; 6: 481-90.
    • (2002) Crit Care , vol.6 , pp. 481-490
    • Haddad, J.J.1
  • 260
    • 0026970404 scopus 로고    scopus 로고
    • Schreck R, Albermann K, Baeuerle PA. Nuclear factor κB: An. oxidative stress responsive transcription factor of eukaryotic cells (a review). Free Radic Res Commun 1992; 17: 221-37.
    • Schreck R, Albermann K, Baeuerle PA. Nuclear factor κB: An. oxidative stress responsive transcription factor of eukaryotic cells (a review). Free Radic Res Commun 1992; 17: 221-37.
  • 262
    • 0033163393 scopus 로고    scopus 로고
    • Li NX, Karin M. Is NF-κB the sensor. of oxidative stress? FASEB J 1999; 13: 1137-43.
    • Li NX, Karin M. Is NF-κB the sensor. of oxidative stress? FASEB J 1999; 13: 1137-43.
  • 263
    • 0033990345 scopus 로고    scopus 로고
    • Oxidative stress and nuclear factor-κB activation. A reassessment of the evidence in the light of recent discoveries
    • Bowie A. O'Neill LAJ. Oxidative stress and nuclear factor-κB activation. A reassessment of the evidence in the light of recent discoveries. Biochem, Pharmacol. 2000; 59: 13-23.
    • (2000) Biochem, Pharmacol , vol.59 , pp. 13-23
    • Bowie, A.1    O'Neill, L.A.J.2
  • 264
    • 33750523632 scopus 로고    scopus 로고
    • NF-κB activation by reactive oxygen species: Fifteen years later
    • Gloire B, Legrand-Poels S, Piette J. NF-κB activation by reactive oxygen species: Fifteen years later. Biochem. Pharmacol 2006; 72: 1493-1505.
    • (2006) Biochem. Pharmacol , vol.72 , pp. 1493-1505
    • Gloire, B.1    Legrand-Poels, S.2    Piette, J.3
  • 265
    • 33750471197 scopus 로고    scopus 로고
    • Mutual cross-talk between reactive oxygen species and nuclear factor-κB: Molecular basis and biological significance
    • Bubici C, Papa S, Dean K, Franzoso G. Mutual cross-talk between reactive oxygen species and nuclear factor-κB: Molecular basis and biological significance. Oncogene 2006; 25: 6731-48.
    • (2006) Oncogene , vol.25 , pp. 6731-6748
    • Bubici, C.1    Papa, S.2    Dean, K.3    Franzoso, G.4
  • 266
    • 0033636352 scopus 로고    scopus 로고
    • Trace elements in regulation of NF-κB activity
    • Kudrin AV. Trace elements in regulation of NF-κB activity. J Trace Elem Med Biol 2000; 14: 129-42.
    • (2000) J Trace Elem Med Biol , vol.14 , pp. 129-142
    • Kudrin, A.V.1
  • 267
    • 0034120712 scopus 로고    scopus 로고
    • Thiol-reactive metal compounds inhibit NF-κB activation by blocking IκB kinase
    • Jeon KI, Jeong JY, Jue DM. Thiol-reactive metal compounds inhibit NF-κB activation by blocking IκB kinase. J Immunol 2000; 164: 5981-9.
    • (2000) J Immunol , vol.164 , pp. 5981-5989
    • Jeon, K.I.1    Jeong, J.Y.2    Jue, D.M.3
  • 268
    • 0028822184 scopus 로고
    • Cupric ion blocks NF-κB activation through inhibiting the signal-induced phosphorylation of IκBα
    • Satake H, Suzuki K, Aoki T, Otsuka M, Sugiura Y, Yamamoto T, et al. Cupric ion blocks NF-κB activation through inhibiting the signal-induced phosphorylation of IκBα. Biochem Biophys Res Commun 1995; 216: 568-73.
    • (1995) Biochem Biophys Res Commun , vol.216 , pp. 568-573
    • Satake, H.1    Suzuki, K.2    Aoki, T.3    Otsuka, M.4    Sugiura, Y.5    Yamamoto, T.6
  • 269
    • 0034675345 scopus 로고    scopus 로고
    • Pyrrolidine dithiocarbamate inhibits TNF-α-dependent activation of NF-κB by increasing intracellular copper level in human aortic smooth muscle cells
    • Iseki A, Kambe F, Okumura K, Niwata S, Yamamoto R, Hayakawa T, et al. Pyrrolidine dithiocarbamate inhibits TNF-α-dependent activation of NF-κB by increasing intracellular copper level in human aortic smooth muscle cells. Biochem Biophys Res Commun 2000; 276: 88-92.
    • (2000) Biochem Biophys Res Commun , vol.276 , pp. 88-92
    • Iseki, A.1    Kambe, F.2    Okumura, K.3    Niwata, S.4    Yamamoto, R.5    Hayakawa, T.6
  • 271
  • 272
    • 0032518199 scopus 로고    scopus 로고
    • Inhibition of NF-κB binding to DNA by chromium, cadmium, mercury, zinc, and arsenite in vitro: Evidence of a thiol mechanism
    • Shumilla JA, Wetterhahn KE, Barchowsky A. Inhibition of NF-κB binding to DNA by chromium, cadmium, mercury, zinc, and arsenite in vitro: Evidence of a thiol mechanism. Arch Biochem Biophys 1998; 349:356-62.
    • (1998) Arch Biochem Biophys , vol.349 , pp. 356-362
    • Shumilla, J.A.1    Wetterhahn, K.E.2    Barchowsky, A.3
  • 273
    • 0032908620 scopus 로고    scopus 로고
    • Zinc is requined in pyrrolidine dithiocarbamate inhibition of NF-κB activation
    • Kim CH, Kim JH, Hsu CY, Ahn YS Zinc is requined in pyrrolidine dithiocarbamate inhibition of NF-κB activation. FEBS Lett 1999; 449: 28-32.
    • (1999) FEBS Lett , vol.449 , pp. 28-32
    • Kim, C.H.1    Kim, J.H.2    Hsu, C.Y.3    Ahn, Y.S.4
  • 274
    • 0028311733 scopus 로고
    • Glutathione mercaptides as transport forms of metals
    • Ballatori N. Glutathione mercaptides as transport forms of metals. Adv Pharmacol 1994; 27: 271-98.
    • (1994) Adv Pharmacol , vol.27 , pp. 271-298
    • Ballatori, N.1
  • 275
    • 0030459341 scopus 로고    scopus 로고
    • 2-mercaptoethanol restores the ability of nuclear factor kappa B (NF-κB) to bind DNA in. nuclear extracts from interleukin 1-treated cells incubated with pyrollidine dithiocarbamate (PDTC). Evidence for oxidation of glutathione in the mechanism of inhibition of NF-κB by PDTC
    • Brennan P, ONeill LAJ. 2-mercaptoethanol restores the ability of nuclear factor kappa B (NF-κB) to bind DNA in. nuclear extracts from interleukin 1-treated cells incubated with pyrollidine dithiocarbamate (PDTC). Evidence for oxidation of glutathione in the mechanism of inhibition of NF-κB by PDTC. Biochem J 1996; 320: 975-81.
    • (1996) Biochem J , vol.320 , pp. 975-981
    • Brennan, P.1    ONeill, L.A.J.2
  • 276
    • 0033233191 scopus 로고    scopus 로고
    • The inhibition of NF-κB activation pathways and the induction of apoptosis by dithiocarbamates in T cells are blocked by the glutathione precursor N-acetyl-L-cysteine
    • Fernandez PC, Machado J, Heussler VT, Botteron C, Palmer GH, Dobbelaere DAE. The inhibition of NF-κB activation pathways and the induction of apoptosis by dithiocarbamates in T cells are blocked by the glutathione precursor N-acetyl-L-cysteine. Biol Chem 1999; 380: 1383-94.
    • (1999) Biol Chem , vol.380 , pp. 1383-1394
    • Fernandez, P.C.1    Machado, J.2    Heussler, V.T.3    Botteron, C.4    Palmer, G.H.5    Dobbelaere, D.A.E.6
  • 277
    • 0037226650 scopus 로고    scopus 로고
    • Thiol antioxidant reversal of pyrrolidine dithiocarbamate-induced reciprocal regulation of AP-1 and NF-κB
    • Kim CH, Kim JH, Lee J, Hsu CY, Ahn YS. Thiol antioxidant reversal of pyrrolidine dithiocarbamate-induced reciprocal regulation of AP-1 and NF-κB. Biol Chem 2003; 384: 143-50.
    • (2003) Biol Chem , vol.384 , pp. 143-150
    • Kim, C.H.1    Kim, J.H.2    Lee, J.3    Hsu, C.Y.4    Ahn, Y.S.5
  • 278
    • 0038150155 scopus 로고    scopus 로고
    • Zinc-induced NF-κB inhibition can be modulated by changes in the intracellular metallothionein level
    • Kim CH, Kim JH, Lee J, Ahn YS. Zinc-induced NF-κB inhibition can be modulated by changes in the intracellular metallothionein level. Toxicol Appl Pharmacol 2003; 190: 189-96.
    • (2003) Toxicol Appl Pharmacol , vol.190 , pp. 189-196
    • Kim, C.H.1    Kim, J.H.2    Lee, J.3    Ahn, Y.S.4
  • 280
    • 0037566901 scopus 로고    scopus 로고
    • For whom the bell tolls: Protein quality control of the endoplasmic reticulum and the ubiquitin-proteasome connection
    • Kostova Z, Wolf DH. For whom the bell tolls: Protein quality control of the endoplasmic reticulum and the ubiquitin-proteasome connection. EMBO J 2003; 22: 2309-17.
    • (2003) EMBO J , vol.22 , pp. 2309-2317
    • Kostova, Z.1    Wolf, D.H.2
  • 281
    • 23944471680 scopus 로고    scopus 로고
    • The ubiquitin system for protein degradation and some of its roles in the control of the cell division cycle
    • Hershko A. The ubiquitin system for protein degradation and some of its roles in the control of the cell division cycle. Cell Death Differ 2005; 12: 1191-7.
    • (2005) Cell Death Differ , vol.12 , pp. 1191-1197
    • Hershko, A.1
  • 282
    • 0141645624 scopus 로고    scopus 로고
    • The role of the ubiquitin/proteasome system in cellular responses to radiation
    • McBride WH, Iwamoto KS, Syljuasen R, Pervan M, Pajonk F. The role of the ubiquitin/proteasome system in cellular responses to radiation. Oncogene 2003; 22: 5755-73.
    • (2003) Oncogene , vol.22 , pp. 5755-5773
    • McBride, W.H.1    Iwamoto, K.S.2    Syljuasen, R.3    Pervan, M.4    Pajonk, F.5
  • 283
    • 0347867373 scopus 로고    scopus 로고
    • Degradation of paternal mitochondria after fertilization: Implications for heteroplasmy, assisted reproductive technologies and mtDNA inheritance
    • Sutovsky P, Van Leyen K, McCauley T, Day BN, Sutovsky M. Degradation of paternal mitochondria after fertilization: Implications for heteroplasmy, assisted reproductive technologies and mtDNA inheritance. Reprod Biomed Online 2004; 8: 24-33.
    • (2004) Reprod Biomed Online , vol.8 , pp. 24-33
    • Sutovsky, P.1    Van Leyen, K.2    McCauley, T.3    Day, B.N.4    Sutovsky, M.5
  • 284
    • 0141729263 scopus 로고    scopus 로고
    • Diverse roles for ubiquitin-dependent proteolysis in transcriptional activation
    • Lipford JR, Deshaies RJ. Diverse roles for ubiquitin-dependent proteolysis in transcriptional activation. Nat Cell Biol 2003; 5: 845-50.
    • (2003) Nat Cell Biol , vol.5 , pp. 845-850
    • Lipford, J.R.1    Deshaies, R.J.2
  • 285
    • 0037335034 scopus 로고    scopus 로고
    • How the ubiquitin-proteasome system controls transcription
    • Muratani M, Tansey WP. How the ubiquitin-proteasome system controls transcription. Nat Rev Mol Cell Biol 2003; 4: 192-201.
    • (2003) Nat Rev Mol Cell Biol , vol.4 , pp. 192-201
    • Muratani, M.1    Tansey, W.P.2
  • 286
    • 33846658069 scopus 로고    scopus 로고
    • The proteasome regulates HIV-1 transcription by both proteolytic and nonproteolytic mechanisms
    • Lassot I, Latreille D, Rousset E, Sourisseau M, Linares LK, Chable-Bessia C, et al. The proteasome regulates HIV-1 transcription by both proteolytic and nonproteolytic mechanisms. Mol Cell 2007; 25: 369-83.
    • (2007) Mol Cell , vol.25 , pp. 369-383
    • Lassot, I.1    Latreille, D.2    Rousset, E.3    Sourisseau, M.4    Linares, L.K.5    Chable-Bessia, C.6
  • 287
    • 0036000003 scopus 로고    scopus 로고
    • Sum1, a component of the fission yeast eIF3 translation initiation complex, is rapidly relocalized during environmental stress and interacts with components of the 26S proteasome
    • Dunand-Sauthier I, Walker C, Wilkinson C, Gordon C, Crane R, Norbury C, et al. Sum1, a component of the fission yeast eIF3 translation initiation complex, is rapidly relocalized during environmental stress and interacts with components of the 26S proteasome. Mol Biol Cell 2002; 13: 1626-40.
    • (2002) Mol Biol Cell , vol.13 , pp. 1626-1640
    • Dunand-Sauthier, I.1    Walker, C.2    Wilkinson, C.3    Gordon, C.4    Crane, R.5    Norbury, C.6
  • 289
    • 27544486193 scopus 로고    scopus 로고
    • The proteasome regulatory particle alters the SAGA coactivator to enhance its interactions with transcriptional activators
    • Lee D, Ezhkova E, Li B, Pattenden SG, Tansey WP, Workman JL. The proteasome regulatory particle alters the SAGA coactivator to enhance its interactions with transcriptional activators. Cell 2005; 123: 423-36.
    • (2005) Cell , vol.123 , pp. 423-436
    • Lee, D.1    Ezhkova, E.2    Li, B.3    Pattenden, S.G.4    Tansey, W.P.5    Workman, J.L.6
  • 290
    • 0035947238 scopus 로고    scopus 로고
    • The 19S regulatory particle of the proteasome is required for efficient transcription elongation by RNA polymerase II
    • Ferdous A, Gonzalez F, Sun L, Kodadek T, Johnston SA. The 19S regulatory particle of the proteasome is required for efficient transcription elongation by RNA polymerase II. Mol Cell 2001; 7: 981-91.
    • (2001) Mol Cell , vol.7 , pp. 981-991
    • Ferdous, A.1    Gonzalez, F.2    Sun, L.3    Kodadek, T.4    Johnston, S.A.5
  • 291
    • 0037134015 scopus 로고    scopus 로고
    • Recruitment of a 19S proteasome subcomplex to an activated promoter
    • Gonzalez F, Delahodde A, Kodadek T, Johnston SA. Recruitment of a 19S proteasome subcomplex to an activated promoter. Science 2002; 296: 548-50.
    • (2002) Science , vol.296 , pp. 548-550
    • Gonzalez, F.1    Delahodde, A.2    Kodadek, T.3    Johnston, S.A.4
  • 292
    • 0034665742 scopus 로고    scopus 로고
    • A nonproteolytic function of the proteasome is required for the dissociation of Cdc2 and cyclin B at the end of M phase
    • Nishiyama A, Tachibana K, Igarashi Y, Yasuda H, Tanahashi N, Tanaka K, et al. A nonproteolytic function of the proteasome is required for the dissociation of Cdc2 and cyclin B at the end of M phase. Genes Dev 2000; 14: 2344-57.
    • (2000) Genes Dev , vol.14 , pp. 2344-2357
    • Nishiyama, A.1    Tachibana, K.2    Igarashi, Y.3    Yasuda, H.4    Tanahashi, N.5    Tanaka, K.6
  • 293
    • 33847346002 scopus 로고    scopus 로고
    • Management of multiple myeloma with bortezomib: Experts review the data and debate the issues
    • Dicato M, Boccadoro M, Cavenagh J, Harousseau JL, Ludwig H, San Miguel J, et al. Management of multiple myeloma with bortezomib: Experts review the data and debate the issues. Oncology 2006; 70: 474-82.
    • (2006) Oncology , vol.70 , pp. 474-482
    • Dicato, M.1    Boccadoro, M.2    Cavenagh, J.3    Harousseau, J.L.4    Ludwig, H.5    San Miguel, J.6
  • 294
    • 0034864799 scopus 로고    scopus 로고
    • Proteasome inhibitors: From research tools to drug candidates
    • Kisselev AF, Goldberg AL. Proteasome inhibitors: From research tools to drug candidates. Chem Biol 2001; 8: 739-58.
    • (2001) Chem Biol , vol.8 , pp. 739-758
    • Kisselev, A.F.1    Goldberg, A.L.2
  • 295
    • 2342667387 scopus 로고    scopus 로고
    • The development of proteasome inhibitors as anticancer drugs
    • Adams J. The development of proteasome inhibitors as anticancer drugs. Cancer Cell 2004; 5: 417-21.
    • (2004) Cancer Cell , vol.5 , pp. 417-421
    • Adams, J.1
  • 296
    • 2342613652 scopus 로고    scopus 로고
    • The proteasome: A suitable antineoplastic target
    • Adams J. The proteasome: A suitable antineoplastic target. Nat Rev Cancer 2004; 4: 349-60.
    • (2004) Nat Rev Cancer , vol.4 , pp. 349-360
    • Adams, J.1
  • 297
    • 33745674468 scopus 로고    scopus 로고
    • Drug discovery in the ubiquitin-proteasome system
    • Nalepa G, Rolfe M, Harper JW. Drug discovery in the ubiquitin-proteasome system. Nat Rev Drug Discov 2006; 5: 596-613.
    • (2006) Nat Rev Drug Discov , vol.5 , pp. 596-613
    • Nalepa, G.1    Rolfe, M.2    Harper, J.W.3
  • 299
    • 0000946534 scopus 로고
    • Structural studies of metal dithiocarbamates. III. The crystal and molecular structure of zinc diethyldithiocarbamate
    • Bonamico M, Mazzone G, Vaciago A, Zambonelli L. Structural studies of metal dithiocarbamates. III. The crystal and molecular structure of zinc diethyldithiocarbamate. Acta Crystallogr 1965; 19: 898-909.
    • (1965) Acta Crystallogr , vol.19 , pp. 898-909
    • Bonamico, M.1    Mazzone, G.2    Vaciago, A.3    Zambonelli, L.4
  • 300
    • 0037902555 scopus 로고    scopus 로고
    • Melanin as a target for melanoma chemotherapy: Pro-oxidant effect of oxygen and metals on melanoma viability
    • Farmer PJ, Gidanian S, Shahandeh B, Di Bilio AJ, Tohidian N, Meyskens FL. Melanin as a target for melanoma chemotherapy: Pro-oxidant effect of oxygen and metals on melanoma viability. Pigment Cell Res 2003; 16: 273-9.
    • (2003) Pigment Cell Res , vol.16 , pp. 273-279
    • Farmer, P.J.1    Gidanian, S.2    Shahandeh, B.3    Di Bilio, A.J.4    Tohidian, N.5    Meyskens, F.L.6
  • 301
    • 11144224746 scopus 로고    scopus 로고
    • Disulfiram facilitates intracellular Cu uptake and induces apoptosis in human melanoma cells
    • Cen D, Brayton D, Shahandeh B, Meyskens FL, Farmer PJ. Disulfiram facilitates intracellular Cu uptake and induces apoptosis in human melanoma cells. J Med Chem 2004; 47: 6914-20.
    • (2004) J Med Chem , vol.47 , pp. 6914-6920
    • Cen, D.1    Brayton, D.2    Shahandeh, B.3    Meyskens, F.L.4    Farmer, P.J.5
  • 302
    • 32144433858 scopus 로고    scopus 로고
    • Suppression of survival in human SKBR3 breast carcinoma in response to metal-chelator complexes is preferential for copper-dithiocarbamate
    • Viola-Rhenals M, Rieber MS, Rieber M. Suppression of survival in human SKBR3 breast carcinoma in response to metal-chelator complexes is preferential for copper-dithiocarbamate. Biochem Pharmacol 2006; 71: 722-34.
    • (2006) Biochem Pharmacol , vol.71 , pp. 722-734
    • Viola-Rhenals, M.1    Rieber, M.S.2    Rieber, M.3
  • 303
    • 33751441045 scopus 로고    scopus 로고
    • Pyrrolidine dithiocarbamate exerts anti-proliferative and pro-apoptotic effects in renal cell carcinoma cell lines
    • Morais C, Pat B, Gobe G, Johnson DW, Healy H. Pyrrolidine dithiocarbamate exerts anti-proliferative and pro-apoptotic effects in renal cell carcinoma cell lines. Nephrol Dial Transplant 2006; 21: 3377-88.
    • (2006) Nephrol Dial Transplant , vol.21 , pp. 3377-3388
    • Morais, C.1    Pat, B.2    Gobe, G.3    Johnson, D.W.4    Healy, H.5
  • 304
    • 33845304732 scopus 로고    scopus 로고
    • Pharmacological profiling of disulfiram using human tumor cell lines and human tumor cells from patiens
    • Wickstrom M, Danielsson K, Rickardson L, Gullbo J, Nygren P, Isaksson A, et al. Pharmacological profiling of disulfiram using human tumor cell lines and human tumor cells from patiens. Biochem Pharmacol 2007; 73: 25-33.
    • (2007) Biochem Pharmacol , vol.73 , pp. 25-33
    • Wickstrom, M.1    Danielsson, K.2    Rickardson, L.3    Gullbo, J.4    Nygren, P.5    Isaksson, A.6
  • 305
    • 4644235752 scopus 로고    scopus 로고
    • Disulfiram inhibits activating transcription factor/cyclic AMP-responsive element binding protein and human melanoma growth in a metal-dependent manner in vitro, in mice and in a patient with metastatic disease
    • Brar SS, Grigg C, Wilson KS, Holder WD, Dreau D, Austin C, et al. Disulfiram inhibits activating transcription factor/cyclic AMP-responsive element binding protein and human melanoma growth in a metal-dependent manner in vitro, in mice and in a patient with metastatic disease. Mol Cancer Ther 2004; 3: 1049-60.
    • (2004) Mol Cancer Ther , vol.3 , pp. 1049-1060
    • Brar, S.S.1    Grigg, C.2    Wilson, K.S.3    Holder, W.D.4    Dreau, D.5    Austin, C.6
  • 306
    • 20744441360 scopus 로고    scopus 로고
    • Pyrrolidine dithiocarbamate reduces coxsackievirus B3 replication through inhibition of the ubiquitin-proteasome pathway
    • Si X, McManus BM, Zhang JC, Yuan J, Cheung C, Esfandiarei M, et al. Pyrrolidine dithiocarbamate reduces coxsackievirus B3 replication through inhibition of the ubiquitin-proteasome pathway. J Virol 2005; 79: 8014-23.
    • (2005) J Virol , vol.79 , pp. 8014-8023
    • Si, X.1    McManus, B.M.2    Zhang, J.C.3    Yuan, J.4    Cheung, C.5    Esfandiarei, M.6
  • 307
    • 27644479061 scopus 로고    scopus 로고
    • Inhibition of polyprotein processing and RNA replication of human rhinovirus by pyrrolidine dithiocarbamate involves metal ions
    • Krenn BM, Holzer B, Gaudemak E, Triendl A, van Kuppeveld FJ, Seipelt J. Inhibition of polyprotein processing and RNA replication of human rhinovirus by pyrrolidine dithiocarbamate involves metal ions. J Virol 2005; 79: 13892-9.
    • (2005) J Virol , vol.79 , pp. 13892-13899
    • Krenn, B.M.1    Holzer, B.2    Gaudemak, E.3    Triendl, A.4    van Kuppeveld, F.J.5    Seipelt, J.6
  • 308
    • 0036932955 scopus 로고    scopus 로고
    • Effect of neurotoxic metal ions on the proteolytic activities of the 20S proteasome ftom bovine brain
    • Amici M, Forti K, Nobili C, Lupidi G, Angeletti M, Fioretti E, et al. Effect of neurotoxic metal ions on the proteolytic activities of the 20S proteasome ftom bovine brain. J Biol Inorg Chem 2002; 7: 750-6.
    • (2002) J Biol Inorg Chem , vol.7 , pp. 750-756
    • Amici, M.1    Forti, K.2    Nobili, C.3    Lupidi, G.4    Angeletti, M.5    Fioretti, E.6
  • 310
    • 3042745312 scopus 로고    scopus 로고
    • Pyrrolidine dithiocarbamate and zinc inhibit proteasome-dependent proteolysis
    • Kim I, Kim CH, Kim JH, Lee J, Choi JJ, Chen ZA, et al. Pyrrolidine dithiocarbamate and zinc inhibit proteasome-dependent proteolysis. Exp.Cell Res 2004; 298: 229-38.
    • (2004) Exp.Cell Res , vol.298 , pp. 229-238
    • Kim, I.1    Kim, C.H.2    Kim, J.H.3    Lee, J.4    Choi, J.J.5    Chen, Z.A.6
  • 311
    • 21344440550 scopus 로고    scopus 로고
    • Inhibition of prostate cancer cellular proteasome activity by a pyrrolidine dithiocarbamate-copper complex is associated with suppression of proliferation and induction of apoptosis
    • Chen D, Peng FY, Cui QC, Daniel KG, Orlu S, Liu JG, et al. Inhibition of prostate cancer cellular proteasome activity by a pyrrolidine dithiocarbamate-copper complex is associated with suppression of proliferation and induction of apoptosis. Front Biosci 2005; 10: 2932-9.
    • (2005) Front Biosci , vol.10 , pp. 2932-2939
    • Chen, D.1    Peng, F.Y.2    Cui, Q.C.3    Daniel, K.G.4    Orlu, S.5    Liu, J.G.6
  • 312
    • 84901648175 scopus 로고    scopus 로고
    • Clioquinol and pyrrolidine dithiocarbamate complex with copper to form proteasome inhibitors and apoptosis inducers in human breast cancer cells
    • Daniel KG, Chen D, Orlu S, Cui QC, Miller FR, Dou QP. Clioquinol and pyrrolidine dithiocarbamate complex with copper to form proteasome inhibitors and apoptosis inducers in human breast cancer cells. Breast Cancer Res 2005; 7: R897-908.
    • (2005) Breast Cancer Res , vol.7
    • Daniel, K.G.1    Chen, D.2    Orlu, S.3    Cui, Q.C.4    Miller, F.R.5    Dou, Q.P.6
  • 313
    • 33751285781 scopus 로고    scopus 로고
    • Disulfiram, a clinically used anti-alcoholism drug and copper-binding agent induces apoptotic cell death in breast cancer cultures and xenografts via inhibition of the proteasome activity
    • Chen D, Cui QC, Yang H, Dou QP. Disulfiram, a clinically used anti-alcoholism drug and copper-binding agent induces apoptotic cell death in breast cancer cultures and xenografts via inhibition of the proteasome activity. Cancer Res 2006; 66: 10425-33.
    • (2006) Cancer Res , vol.66 , pp. 10425-10433
    • Chen, D.1    Cui, Q.C.2    Yang, H.3    Dou, Q.P.4
  • 315
    • 14944384045 scopus 로고    scopus 로고
    • Synthesis, characterization, and comparative in vitro cytotoxicity studies of platinum(II), palladium(II), and gold(III) methylsarcosinedithiocarbamate complexes
    • Giovagnini L, Ronconi L, Aldinucci D, Lorenzon D, Sitran S, Fregona D. Synthesis, characterization, and comparative in vitro cytotoxicity studies of platinum(II), palladium(II), and gold(III) methylsarcosinedithiocarbamate complexes. J Med Chem 2005; 48: 1589-95.
    • (2005) J Med Chem , vol.48 , pp. 1589-1595
    • Giovagnini, L.1    Ronconi, L.2    Aldinucci, D.3    Lorenzon, D.4    Sitran, S.5    Fregona, D.6
  • 316
    • 33644855554 scopus 로고    scopus 로고
    • Gold(III) dithiocarbamate derivatives for the treatment of cancer: Solution chemistry, DNA binding, and hemolytic properties
    • Ronconi L, Marzano C, Zanello P, Corsini M, Miolo G, Macca C, et al. Gold(III) dithiocarbamate derivatives for the treatment of cancer: Solution chemistry, DNA binding, and hemolytic properties. J Med Chem 2006; 49: 1648-57.
    • (2006) J Med Chem , vol.49 , pp. 1648-1657
    • Ronconi, L.1    Marzano, C.2    Zanello, P.3    Corsini, M.4    Miolo, G.5    Macca, C.6
  • 317
    • 1542270307 scopus 로고    scopus 로고
    • Organic copper complexes as a new class of proteasome inhibitors and apoptosis inducers in human cancer cells
    • Daniel KG, Gupta P, Harbach RH, Guida WC, Dou QP. Organic copper complexes as a new class of proteasome inhibitors and apoptosis inducers in human cancer cells. Biochem Pharmacol 2004; 67: 1139-51.
    • (2004) Biochem Pharmacol , vol.67 , pp. 1139-1151
    • Daniel, K.G.1    Gupta, P.2    Harbach, R.H.3    Guida, W.C.4    Dou, Q.P.5
  • 318
    • 33845372349 scopus 로고    scopus 로고
    • Novel Schiff base copper complexes of quinoline-2 carboxaldehyde as proteasome inhibitors in human prostate cancer cells
    • Adsule S, Barve V, Chen D, Ahmed F, Dou QP, Padhye S, et al. Novel Schiff base copper complexes of quinoline-2 carboxaldehyde as proteasome inhibitors in human prostate cancer cells. J Med Chem 2006; 49: 7242-6.
    • (2006) J Med Chem , vol.49 , pp. 7242-7246
    • Adsule, S.1    Barve, V.2    Chen, D.3    Ahmed, F.4    Dou, Q.P.5    Padhye, S.6
  • 319
    • 0037331103 scopus 로고    scopus 로고
    • RING finger ubiquitin protein ligases: Implications for tumorigenesis, metastasis and for molecular targets in cancer
    • Fang S, Lorick KL, Jensen JP, Weissman AM. RING finger ubiquitin protein ligases: Implications for tumorigenesis, metastasis and for molecular targets in cancer. Semin Cancer Biol 2003; 13: 5-14.
    • (2003) Semin Cancer Biol , vol.13 , pp. 5-14
    • Fang, S.1    Lorick, K.L.2    Jensen, J.P.3    Weissman, A.M.4
  • 321
    • 36349036870 scopus 로고    scopus 로고
    • Disulfiram inhibits the E3 ligase activity of breast cancer associated gene 2 (BCA2) and the growth of BCA2-expressing breast cancers in vitro and in vivo
    • Burger AM, Phatak P, Wilson M, Seth AK. Disulfiram inhibits the E3 ligase activity of breast cancer associated gene 2 (BCA2) and the growth of BCA2-expressing breast cancers in vitro and in vivo. Eur J Cancer Suppl. 2006; 4: 118.
    • (2006) Eur J Cancer Suppl , vol.4 , pp. 118
    • Burger, A.M.1    Phatak, P.2    Wilson, M.3    Seth, A.K.4
  • 323
    • 19344364762 scopus 로고    scopus 로고
    • JAMM: A metalloprotease-like zinc site in the proteasome and signalosome
    • Ambroggio XI, Rees DC, Deshaies RJ. JAMM: A metalloprotease-like zinc site in the proteasome and signalosome. PLoS Biol 2004; 2: 113-9.
    • (2004) PLoS Biol , vol.2 , pp. 113-119
    • Ambroggio, X.I.1    Rees, D.C.2    Deshaies, R.J.3
  • 324
    • 0037131242 scopus 로고    scopus 로고
    • Role of predicted metalloprotease motif of Jab1/Csn5 in cleavage of Nedd8 from Cull
    • Cope GA, Suh GSB, Aravind L, Schwarz SE, Zipursky SL, Koonin EV, et al. Role of predicted metalloprotease motif of Jab1/Csn5 in cleavage of Nedd8 from Cull. Science 2002; 298: 608-11.
    • (2002) Science , vol.298 , pp. 608-611
    • Cope, G.A.1    Suh, G.S.B.2    Aravind, L.3    Schwarz, S.E.4    Zipursky, S.L.5    Koonin, E.V.6
  • 325
    • 0037131243 scopus 로고    scopus 로고
    • Role of Rpn11 metalloprotease in deubiquitination and degradation by the 26S proteasome
    • Verma R, Aravind L, Oania R, McDonald WH, Yates JR, Koonin EV, et al. Role of Rpn11 metalloprotease in deubiquitination and degradation by the 26S proteasome. Science 2002; 298: 611-5.
    • (2002) Science , vol.298 , pp. 611-615
    • Verma, R.1    Aravind, L.2    Oania, R.3    McDonald, W.H.4    Yates, J.R.5    Koonin, E.V.6
  • 326
    • 7744244599 scopus 로고    scopus 로고
    • Recent advances in zinc enzymology
    • Lipscomb WN, Strater N. Recent advances in zinc enzymology. Chem Rev 1996; 96: 2375-433.
    • (1996) Chem Rev , vol.96 , pp. 2375-2433
    • Lipscomb, W.N.1    Strater, N.2
  • 327
    • 0142148099 scopus 로고    scopus 로고
    • Inhibition of invasion and angiogenesis by zinc-chelating agent disulfiram
    • Shiah SG, Kao YR, Wu FYH, Wu CW. Inhibition of invasion and angiogenesis by zinc-chelating agent disulfiram. Mol Pharmacol 2003; 64: 1076-84
    • (2003) Mol Pharmacol , vol.64 , pp. 1076-1084
    • Shiah, S.G.1    Kao, Y.R.2    Wu, F.Y.H.3    Wu, C.W.4
  • 328
    • 0027269781 scopus 로고
    • 2 and antioxidants have opposite effects on activation of NF-κB and AP-1 in intact cells: AP-1 as secondary antioxidant-responsive factor
    • 2 and antioxidants have opposite effects on activation of NF-κB and AP-1 in intact cells: AP-1 as secondary antioxidant-responsive factor. EMBO J 1993; 12: 2005-15.
    • (1993) EMBO J , vol.12 , pp. 2005-2015
    • Meyer, M.1    Schreck, R.2    Baeuerle, P.A.3
  • 329
    • 0031728648 scopus 로고    scopus 로고
    • Transcriptional regulation of the heme oxygenase 1 gene by pyrrolidine dithiocarbamate
    • Hartsfield CL, Alam J, Choi AMK. Transcriptional regulation of the heme oxygenase 1 gene by pyrrolidine dithiocarbamate. FASEB J 1998; 12: 1675-82.
    • (1998) FASEB J , vol.12 , pp. 1675-1682
    • Hartsfield, C.L.1    Alam, J.2    Choi, A.M.K.3
  • 330
    • 0031574131 scopus 로고    scopus 로고
    • NF-κB-independent transcriptional induction of the human manganous superoxide dismutase gene
    • Borrello S, Demple B. NF-κB-independent transcriptional induction of the human manganous superoxide dismutase gene. Arch Biochem Biophys 1997; 348: 289-94.
    • (1997) Arch Biochem Biophys , vol.348 , pp. 289-294
    • Borrello, S.1    Demple, B.2
  • 331
    • 2542626623 scopus 로고    scopus 로고
    • Proteasome inhibitors up-regulate haem oxygenase-1 gene expression: Requirement of p38 MAPK (mitogen-activated protein kinase) activation but not of NF-κB (nuclear factor κB) inhibition
    • Wu WT, Chi KH, Ho FM, Tsao WC, Lin WW. Proteasome inhibitors up-regulate haem oxygenase-1 gene expression: Requirement of p38 MAPK (mitogen-activated protein kinase) activation but not of NF-κB (nuclear factor κB) inhibition. Biochem J 2004; 379: 587-93.
    • (2004) Biochem J , vol.379 , pp. 587-593
    • Wu, W.T.1    Chi, K.H.2    Ho, F.M.3    Tsao, W.C.4    Lin, W.W.5
  • 332
    • 33744947551 scopus 로고    scopus 로고
    • The 19 S proteasomal subunit POH1 contributes to the regulation of c-Jun ubiquitination, stability, and sub-cellular localization
    • Nabhan JF, Ribeiro P. The 19 S proteasomal subunit POH1 contributes to the regulation of c-Jun ubiquitination, stability, and sub-cellular localization. J Biol Chem 2006; 281: 16099-107.
    • (2006) J Biol Chem , vol.281 , pp. 16099-16107
    • Nabhan, J.F.1    Ribeiro, P.2
  • 333
    • 33947255054 scopus 로고    scopus 로고
    • Apoptosis induced by proteasome inhibition in cancer cells: Predominant role of the p53/PUMA pathway
    • Concannon CG, Koehler BF, Reimertz C, Murphy BM, Bonner C, Thurow N, et al. Apoptosis induced by proteasome inhibition in cancer cells: predominant role of the p53/PUMA pathway. Oncogene 2007; 26: 1681-92.
    • (2007) Oncogene , vol.26 , pp. 1681-1692
    • Concannon, C.G.1    Koehler, B.F.2    Reimertz, C.3    Murphy, B.M.4    Bonner, C.5    Thurow, N.6
  • 334
    • 0036683674 scopus 로고    scopus 로고
    • Copper uptake is required for pyrrolidine dithiocarbamate-mediated oxidation and protein level increase of p53 in cells
    • Furuta S, Ortiz F, Sun XZ, Wu HH, Mason A, Momand J. Copper uptake is required for pyrrolidine dithiocarbamate-mediated oxidation and protein level increase of p53 in cells. Biochem J 2002; 365: 639-48.
    • (2002) Biochem J , vol.365 , pp. 639-648
    • Furuta, S.1    Ortiz, F.2    Sun, X.Z.3    Wu, H.H.4    Mason, A.5    Momand, J.6
  • 335
    • 33847689863 scopus 로고    scopus 로고
    • Jab1 as a mediator of nuclear export and cytoplasmic degradation of p53
    • Lee EW, Oh W, Song J. Jab1 as a mediator of nuclear export and cytoplasmic degradation of p53. Mol Cells 2006; 22: 133-40.
    • (2006) Mol Cells , vol.22 , pp. 133-140
    • Lee, E.W.1    Oh, W.2    Song, J.3
  • 336
    • 14544269951 scopus 로고    scopus 로고
    • NF-κB in human disease: Current inhibitors and prospects for de novo structure based design of inhibitors
    • Pande V, Ramos MJ. NF-κB in human disease: Current inhibitors and prospects for de novo structure based design of inhibitors. Curr Med Chem 2005; 12: 357-74.
    • (2005) Curr Med Chem , vol.12 , pp. 357-374
    • Pande, V.1    Ramos, M.J.2
  • 338
    • 33644817495 scopus 로고    scopus 로고
    • Pyridine N-oxide derivatives inhibit viral transactivation by interfering with NF-κB binding
    • Stevens M, Pannecouque C, De Clercq E, Balzarini J. Pyridine N-oxide derivatives inhibit viral transactivation by interfering with NF-κB binding. Biochem Pharmacol 2006; 71: 1122-35.
    • (2006) Biochem Pharmacol , vol.71 , pp. 1122-1135
    • Stevens, M.1    Pannecouque, C.2    De Clercq, E.3    Balzarini, J.4
  • 339
    • 33750446341 scopus 로고    scopus 로고
    • Inhibitors of NF-κB signaling: 785 and counting
    • Gilmore TD, Herscovitch M. Inhibitors of NF-κB signaling: 785 and counting. Oncogene 2006; 25: 6887-99.
    • (2006) Oncogene , vol.25 , pp. 6887-6899
    • Gilmore, T.D.1    Herscovitch, M.2
  • 340
    • 33746862576 scopus 로고    scopus 로고
    • Naturally occurring NF-κB inhibitors
    • Nam NH. Naturally occurring NF-κB inhibitors. Mini Rev Med Chem 2006; 6: 945-51.
    • (2006) Mini Rev Med Chem , vol.6 , pp. 945-951
    • Nam, N.H.1
  • 341
    • 33847188791 scopus 로고    scopus 로고
    • NF-κB inhibitors for the treatment of inflammatory diseases and cancer
    • Calzado MA, Bacher S, Schmitz ML. NF-κB inhibitors for the treatment of inflammatory diseases and cancer. Curr Med Chem 2007; 14: 367-76.
    • (2007) Curr Med Chem , vol.14 , pp. 367-376
    • Calzado, M.A.1    Bacher, S.2    Schmitz, M.L.3
  • 343
    • 1842870662 scopus 로고    scopus 로고
    • In vitro toxicity of several dithiocarbamates and structure-activity relationships
    • Segovia N, Crovetto G, Lardelli P, Espigares M. In vitro toxicity of several dithiocarbamates and structure-activity relationships. J Appl Toxicol 2002; 22: 353-7.
    • (2002) J Appl Toxicol , vol.22 , pp. 353-357
    • Segovia, N.1    Crovetto, G.2    Lardelli, P.3    Espigares, M.4
  • 344
    • 33751275548 scopus 로고    scopus 로고
    • A novel anticancer gold(III) dithiocarbamate compound inhibits the activity of a purified 20S proteasome and 26S proteasome in human breast cancer cell cultures and xenografts
    • Milacic V, Chen D, Ronconi L, Landis-Piwowar KR, Fregona D, Dou QP. A novel anticancer gold(III) dithiocarbamate compound inhibits the activity of a purified 20S proteasome and 26S proteasome in human breast cancer cell cultures and xenografts. Cancer Res 2006; 66: 10478-86.
    • (2006) Cancer Res , vol.66 , pp. 10478-10486
    • Milacic, V.1    Chen, D.2    Ronconi, L.3    Landis-Piwowar, K.R.4    Fregona, D.5    Dou, Q.P.6


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.