252
Views
55
CrossRef citations to date
0
Altmetric
Research Article

GLUTATHIONE CONJUGATES AND THEIR SYNTHETIC DERIVATIVES AS INHIBITORS OF GLUTATHIONE-DEPENDENT ENZYMES INVOLVED IN CANCER AND DRUG RESISTANCE

&
Pages 821-863 | Published online: 27 Nov 2002

REFERENCES

  • Shi Z.Z., Osei F.J., Kala G., Kala S.V., Barrios R.J., Habib G.M., Lukin D.J., Danney C.M., Matzuk M.M., Lieberman M.W. Glutathione Synthesis Is Essential for Mouse Development But Not for Cell Growth in Culture. Proc. Natl Acad. Sci. USA 2000; 97(10)5101–5106
  • Davis W., Jr., Ronai Z., Tew K.D. Cellular Thiols and Reactive Oxygen Species in Drug-Induced Apoptosis. J. Pharmacol. Exp. Ther. 2001; 296(1)1–6
  • Orlowski M., Meister A. The γ-Glutamyl Cycle—A Possible Transport System for Amino Acids. Proc. Natl Acad. Sci. USA 1970; 67: 1248–1255
  • Meister A., Larsson A. Glutathione Synthetase Deficiency and Other Disorders of the γ-Glutamyl Cycle. The Metabolic and Molecular Bases of Inherited Disease, C.F. Scriver, A.L. Beaudet, W.S. Sly, D. Valle. McGraw Hill, New York 1995; 1461–1477
  • Ristoff E., Larsson A. Patients with Genetic Defects in the Gamma-Glutamyl Cycle. Chem. Biol. Interact. 1998; 111–112: 113–121
  • Hammond C.L., Lee T.K., Ballatori N. Novel Roles for Glutathione in Gene Expression, Cell Death, and Membrane Transport of Organic Solutes. J. Hepatol. 2001; 34(6)946–954
  • Locigno R., Castronovo V. Reduced Glutathione System: Role in Cancer Development, Prevention and Treatment. Int. J. Oncol. 2001; 19(2)221–236
  • Hayes J.D., McLellan L.I. Glutathione and Glutathione-Dependent Enzymes Represent a Co-ordinately Regulated Defence Against Oxidative Stress. Free Radic. Res. 1999; 31(4)273–300
  • Baillie T.A., Kassahum K. Reversibility in Glutathione-Conjugate Formation. Adv. Pharmacol. 1994; 27: 163–181
  • Diah S.K., Smitherman P.K., Townsend A.J., Morrow C.S. Detoxification of 1-Chloro-2,4-dinitrobenzene in MCF7 Breast Cancer Cells Expressing Glutathione S-Transferase P1-1 and/or Multidrug Resistance Protein 1. Toxicol. Appl. Pharmacol. 1999; 157(2)85–93
  • Morrow C.S., Smitherman P.K., Townsend A.J. Combined Expression of Multidrug Resistance Protein (MRP) and Glutathione S-Transferase P1-1(GSTP1-1) in MCF7 Cells and High Level Resistance to the Cytotoxicities of Ethacrynic Acid But Not Oxazaphosphorines or Cisplatin. Biochem. Pharmacol. 1998; 56(8)1013–1021
  • Morrow C.S., Smitherman P.K., Diah S.K., Schneider E., Townsend A.J. Coordinated Action of Glutathione S-Transferases (GSTs) and Multidrug Resistance Protein 1(MRP1) in Antineoplastic Drug Detoxification. Mechanism of GST A1-1- and MRP1-Associated Resistance to Chlorambucil in MCF7 Breast Carcinoma Cells. J. Biol. Chem. 1998; 273(32)20114–20120
  • Morrow C.S., Smitherman P.K., Townsend A.J. Role of Multidrug-Resistance Protein 2 in Glutathione S-Transferase P1-1-Mediated Resistance to 4-Nitroquinoline 1-Oxide Toxicities in HepG2 Cells. Mol. Carcinog. 2000; 29: 170–178
  • Paumi C.M., Ledford B.G., Smitherman P.K., Townsend A.J., Morrow C.S. Role of Multidrug Resistance Protein 1 (MRP1) and Glutathione S-Transferase A1-1 in Alkylating Agent Resistance. Kinetics of Glutathione Conjugate Formation and Efflux Govern Differential Cellular Sensitivity to Chlorambucil Versus Melphalan Toxicity. J. Biol. Chem. 2001; 276(11)7952–7956
  • Townsend A.J., Fields W.R., Haynes R.L., Doss A.J., Li Y., Doehmer J., Morrow C.S. Chemoprotective Functions of Glutathione S-Transferases in Cell Lines Induced to Express Specific Isozymes by Stable Transfection. Chem. Biol. Interact. 1998; 111–112: 389–407
  • Cnubben N.H., Rommens A.J., Oudshoorn M.J., van Bladeren P.J. Glutathione-Dependent Biotransformation of the Alkylating Drug Thiotepa and Transport of Its Metabolite Monoglutathionylthiotepa in Human MCF-7 Breast Cancer Cells. Cancer Res. 1998; 58(20)4616–4623
  • Ciaccio P.J., Shen H., Jaiswal A.K., Lyttle M.H., Tew K.D. Modulation of Detoxification Gene Expression in Human Colon HT29 Cells by Glutathione-S-transferase Inhibitors. Mol. Pharmacol. 1995; 48(4)639–647
  • Ciaccio P.J., Tew K.D. Adaptive Response to Glutathione S-Transferase Inhibitors. Br. J. Cancer Suppl. 1996; 27: S93–S98
  • Tew K.D., O'Brien M., Laing N.M., Shen H. Coordinate Changes in Expression of Protective Genes in Drug-Resistant Cells. Chem. Biol. Interact. 1998; 111–112: 199–211
  • Rosario L.A., O'Brien M.L., Henderson C.J., Wolf C.R., Tew K.D. Cellular Response to a Glutathione S-Transferase P1-1 Activated Prodrug. Mol. Pharmacol. 2000; 58(1)167–174
  • O'Brien M., Kruh G.D., Tew K.D. The Influence of Coordinate Overexpression of Glutathione Phase II Detoxification Gene Products on Drug Resistance. J. Pharmacol. Exp. Ther. 2000; 294(2)480–487
  • Tatebe S., Sinicrope F.A., Tien K.M. Induction of Multidrug Resistance Proteins MRP1 and MRP3 and [Gamma]-Glutamylcysteine Synthetase Gene Expression by Nonsteroidal Anti-inflammatory Drugs in Human Colon Cancer Cells. Biochem. Biophys. Res. Commun. 2002; 290(5)1427–1433
  • Perquin M., Oster T., Maul A., Froment N., Untereiner M., Bagrel D. The Glutathione-Related Detoxification Pathway in the Human Breast: A Highly Coordinated System Disrupted in the Tumour Tissues. Cancer Lett. 2000; 158(1)7–16
  • Sagara Y., Dargusch R., Chambers D., Davis J., Schubert D., Maher P. Cellular Mechanisms of Resistance to Chronic Oxidative Stress. Free Radic. Biol. Med. 1998; 24(9)1375–1389
  • Lam B.K., Frank A.K. Leukotriene C4 Synthase. A Pivotal Enzyme in the Biosynthesis of the Cysteinyl Leukotrienes. Am. J. Respir. Crit. Care Med. 2000; 161(2 Pt 2)S16–S19
  • Chiuch C.C., Rauhala P. The Redox Pathway of S-Nitrosoglutathione, Glutathione and Nitric Oxide in Cell to Neuron Communications. Free Radic. Res. 1999; 31(6)641–650
  • Monks T.J., Lau S.S. Glutathione Conjugation as Mechanism for the Transport of Reactive Metabolites. Adv. Pharmacol. 1994; 27: 183–210
  • Ruscoe J.E., Rosario L.A., Wang T., Gate L., Arifoglu P., Wolf C.R., Henderson C.J., Ronai Z., Tew K.D. Pharmacologic or Genetic Manipulation of Glutathione S-Transferase P1-1(GSTpi) Influences Cell Proliferation Pathways. J. Pharmacol. Exp. Ther. 2001; 298(1)339–345
  • Lucente G., Luisi G., Pinnen F. Design and Synthesis of Glutathione Analogues. Farmaco 1998; 53(12)721–735
  • Adang A.E., Brussee J., Meyer D.J., Coles B., Ketterer B., van der Gen A., Mulder G.J. Substrate Specificity of Rat Liver Glutathione S-Transferase Isoenzymes for a Series of Glutathione Analogues, Modified at the Gamma-Glutamyl Moiety. Biochem. J. 1988; 255(2)721–724
  • Adang A.E., Brussee J., van der Gen A., Mulder G.J. The Glutathione-Binding Site in Glutathione S-Transferases. Investigation of the Cysteinyl, Glycyl and Gamma-Glutamyl Domains. Biochem. J. 1990; 269(1)47–54
  • Lyttle M.H., Hocker M.D., Hui H.C., Caldwell C.G., Aaron D.T., Engqvist G.A., Flatgaard J.E., Bauer K.E. Isozyme-Specific Glutathione-S-transferase Inhibitors: Design and Synthesis. J. Med. Chem. 1994; 37(1)189–194
  • Flatgaard J.E., Bauer K.E., Kauvar L.M. Isozyme Specificity of Novel Glutathione-S-transferase Inhibitors. Cancer Chemother. Pharmacol. 1993; 33(1)63–70
  • Adang A.E., Brussee J., van der Gen A., Mulder G.J. Inhibition of Rat Liver Glutathione S-Transferase Isoenzymes by Peptides Stabilized Against Degradation by Gamma-Glutamyl Transpeptidase. J. Biol. Chem. 1991; 266(2)830–836
  • Adang A.E., Moree W.J., Brussee J., Mulder G.J., van der Gen A. Inhibition of Glutathione S-Transferase 3-3 by Glutathione Derivatives That Bind Covalently to the Active Site. Biochem. J. 1991; 278(Pt 1)63–68
  • Ouwerkerk-Mahadevan S., Mulder G.J. Inhibition of Glutathione Conjugation in the Rat In Vivo by Analogues of Glutathione Conjugates. Chem. Biol. Interact. 1998; 111–112: 163–176
  • Murthy N.S., Bakeris T., Kavarana M.J., Hamilton D.S., Lan Y., Creighton D.J. S-(N-Aryl-N-Hydroxycarbamoyl)Glutathione Derivatives Are Tight-Binding Inhibitors of Glyoxalase I and Slow Substrates for Glyoxalase II. J. Med. Chem. 1994; 37(14)2161–2166
  • Kavarana M.J., Kovaleva E.G., Creighton D.J., Wollman M.B., Eiseman J.L. Mechanism-Based Competitive Inhibitors of Glyoxalase I: Intracellular Delivery, In Vitro Antitumor Activities, and Stabilities in Human Serum and Mouse Serum. J. Med. Chem. 1999; 42(2)221–228
  • D'-Silva C., Daunes S., Rock P., Yardley V., Croft S.L. Structure–Activity Study on the In Vitro Antiprotozoal Activity of Glutathione Derivatives. J. Med. Chem. 2000; 43(10)2072–2078
  • Daunes S., D'Silva C., Kendrick H., Yardley V., Croft S.L. QSAR Study on the Contribution of Log P and E(s) to the In Vitro Antiprotozoal Activity of Glutathione Derivatives. J. Med. Chem. 2001; 44(18)2976–2983
  • Brophy P.M., Campbell A.M., van Eldik A.J., Teesdale-Spittle P.H., Liebau E., Wang M.F. Beta-Carbonyl Substituted Glutathione Conjugates as Inhibitors of O. volvulus GST2. Bioorg. Med. Chem. Lett. 2000; 10(9)979–981
  • Kahna A.H., Bundgaard H. Synthesis and Chemical and Enzymatic Hydrolysis Kinetics of Various Mono- and Diester Prodrugs of N-Acetyl Cysteine. Int. J. Pharm. 1990; 62: 193–205
  • Hobbs M.J., Butterworth M., Cohen G.M., Upshall D.G. Structure–Activity Relationships of Cysteine Esters and Their Effects on Thiol Levels in Rat Lung In Vitro. Biochem. Pharmacol. 1993; 45(8)1605–1612
  • Levy E.J., Andrews P.A., Meister A. Transport of Glutathione Diethyl Ester into Human Cells. Proc. Natl Acad. Sci. USA 1993; 90: 9171–9175
  • Kunze T. Phosphono Analogues of Glutathione as New Inhibitors of Glutathione S-Transferases. Arch. Pharm. Weinheim 1996; 329(11)503–509
  • Kunze T., Heps S. Phosphono Analogs of Glutathione: Inhibition of Glutathione Transferases, Metabolic Stability, and Uptake by Cancer Cells. Biochem. Pharmacol. 2000; 59(8)973–981
  • Chen W.J., Lee D.Y., Armstrong R.N. N4-(Malonyl-d-cysteinyl)-l-2,4-diaminobutyrate: The End-Group Modified Retro-inverso Isomer of Glutathione. J. Org. Chem. 1986; 51(14)2848–2850
  • Luisi G., Calcagni A., Pinnen F. Ψ(SO2–NH) Transition State Isosteres of Peptides. Synthesis of the Glutathione Disulfide Analogue [Glu-Ψ(SO2–NH)–cys–gly]2. Tetrahedron Lett. 1993; 34(14)2391–2392
  • Calcagni A., Dupre S., Lucente G., Luisi G., Pinnen F., Rossi D. Synthesis and Activity of the Glutathione Analogue Gamma-(l-gamma-azaglutamyl)-l-cysteinyl-glycine. Int. J. Pept. Protein Res. 1995; 46(5)434–439
  • Calcagni A., Dupre S., Lucente G., Luisi G., Pinnen F., Rossi D., Spirito A. Synthesis and Activity of the Glutathione Analogue Gamma-(l-gamma-oxaglutamyl)-l-cysteinyl-glycine. Arch. Pharm. Weinheim 1996; 329(11)498–502
  • Vince R., Brownell J., Akella L.B. Synthesis and Activity of Gamma-(l-gamma-azaglutamyl)-S-(p-bromobenzyl)-l-cysteinylglycine: A Metabolically Stable Inhibitor of Glyoxalase I. Bioorg. Med. Chem. Lett. 1999; 9(6)853–856
  • Burg D., Filippov D.V., Hermanns R., van der Marel G.A., van Boom J.H., Mulder G.J. Peptidomimetic Glutathione Analogues as Novel γGT Stable GST Inhibitors. Bioorg. Med. Chem. 2002; 10(1)195–205
  • Klotz P., Slaoui H.A., Baneres J.L., Duckert J.F., Rossi J.C., Kerbal A. Synthesis and Glutathione S-Transferase Structure–Affinity Relationships of Nonpeptide and Peptidase-Stable Glutathione Analogues. J. Med. Chem. 1998; 41(13)2278–2288
  • Duckert J.F., Balas L., Rossi J.C. Synthesis of 5-(Mercaptomethyl)-3(E)-undecene-1,11-dioic Acid, a Non-peptide Glutathione Analog. Tetrahedron Lett. 2001; 42: 3709–3711
  • Sheehan D., Meade G., Foley V.M., Dowd C.A. Structure, Function and Evolution of Glutathione Transferases—Implications for Classification of Non-mammalian Members of an Ancient Enzyme Superfamily. Biochem. J. 2001; 360: 1–16
  • Whalen R., Boyer T.D. Human Glutathione S-Transferases. Semin. Liver Dis. 1998; 18(4)345–358
  • Hayes J.D., Pulford D.J. The Glutathione S-Transferase Supergene Family: Regulation of GST and the Contribution of the Isoenzymes to Cancer Chemoprotection and Drug Resistance. Crit. Rev. Biochem. Mol. Biol. 1995; 30(6)445–600
  • Board P.G., Coggan M., Chelvanayagam G., Easteal S., Jermiin L.S., Schulte G.K., Danley D.E., Hoth L.R., Griffor M.C., Kamath A.V., Rosner M.H., Chrunyk B.A., Perregaux D.E., Gabel C.A., Geoghegan K.F., Pandit J. Identification, Characterization, and Crystal Structure of the Omega Class Glutathione Transferases. J. Biol. Chem. 2000; 275: 24798–24806
  • Pemble S.E., Wardle A.F., Taylor J.B. Glutathione S-Transferase Class Kappa-Characterization by the Cloning of Rat Mitochondrial GST and Identification of a Human Homologue. Biochem. J. 1996; 319: 749–754
  • Board P.G., Baker R.T., Chelvanayagam G., Jermiin L.S. Zeta, a Novel Class of Glutathione Transferases in a Range of Species from Plants to Human. Biochem. J. 1997; 328: 929–935
  • Jakobsson P.J., Morgenstern R., Mancini J., Ford-Hutchinson A., Persson B. Membrane-Associated Proteins in Eicosanoid and Glutathione Metabolism (MAPEG). A Widespread Protein Superfamily. Am. J. Respir. Crit. Care Med. 2000; 161(2 Pt 2)S20–S24
  • Pettigrew N.E., Colman R.F. Heterodimers of Glutathione S-Transferase Can Form Between Isoenzyme Classes Pi and Mu. Arch. Biochem. Biophys. 2001; 396(2)225–230
  • Gardner J.L., Gallagher E.P. Development of a Peptide Antibody Specific to Human Glutathione S-Transferase Alpha 4-4 (hGSTA4-4) Reveals Preferential Localization in Human Liver Mitochondria. Arch. Biochem. Biophys. 2001; 390(1)19–27
  • Quondamatteo F., Schulz T.G., Bunzel N., Hallier E., Herken R. Immunohistochemical Localization of Glutathione S-Transferase-T1 in Murine Kidney, Liver, and Lung. Histochem. Cell Biol. 1998; 110(4)417–423
  • Landi S. Mammalian Class Theta GST and Differential Susceptibility to Carcinogens: A Review. Mutat. Res. 2000; 463(3)247–283
  • Ali-Osman F., Brunner J.M., Kutluk T.M., Hess K. Prognostic Significance of Glutathione S-Transferase Pi Expression and Subcellular Localization in Human Gliomas. Clin. Cancer Res. 1997; 3(12 Pt 1)2253–2261
  • van Lieshout E.M., van Haelst U.J., Wobbes T., Peters W.H. Immunohistochemical Localization of Glutathione S-Transferase Alpha and Pi in Human Esophageal Squamous Epithelium, Barrett's Epithelium and Carcinoma. Jpn. J. Cancer Res. 1999; 90(5)530–535
  • Chen Y.K., Lin L.M. Immunohistochemical Demonstration of Epithelial Glutathione S-Transferase Isoenzymes in Normal, Benign, Premalignant and Malignant Human Oral Mucosa. J. Oral Pathol. Med. 1995; 24(7)316–321
  • Goto S., Ihara Y., Urata Y., Izumi S., Abe K., Koji T., Kondo T. Doxorubicin-Induced DNA Intercalation and Scavenging by Nuclear Glutathione S-Transferase Pi. FASEB J. 2001; 15(14)2702–2714
  • Yu L.G., Fernig D.G., White M.R., Spiller D.G., Appleton P., Evans R.C., Grierson I., Smith J.A., Davies H., Gerasimenko O.V., Petersen O.H., Milton J.D., Rhodes J.M. Edible Mushroom (Agaricus bisporus) Lectin, Which Reversibly Inhibits Epithelial Cell Proliferation, Blocks Nuclear Localization Sequence-Dependent Nuclear Protein Import. J. Biol. Chem. 1999; 274(8)4890–4899
  • Armstrong R.N. Structure, Catalytic Mechanism, and Evolution of the Glutathione Transferases. Chem. Res. Toxicol. 1997; 10(1)2–18
  • Boyer T.D., Vessay D.A., Holcomb C., Saley N. Studies of the Relationship Between the Catalytic Activity and Binding of Nonsubstrate Ligands by Glutathione S-Transferases. Biochem. J. 1984; 217: 179–185
  • Listowski I., Abramovitz M., Homma H., Niitsu Y. Intracellular Binding and Transport of Hormones and Xenobiotics by Glutathione S-Transferases. Drug Metab. Rev. 1988; 19: 305–318
  • Mannervik B., Danielson U.H. Glutathione Transferases—Structure and Catalytic Activity. CRC Crit. Rev. Biochem. 1988; 23(3)283–337
  • Oakley A.J., LoBello M., Nuccetelli M., Mazzetti A.P., Parker M.W. The Ligandin (Non-substrate) Binding Site of Human Pi Class Glutathione Transferase Is Located in the Electrophile Binding Site (H-Site). J. Mol. Biol. 1999; 291(4)913–926
  • Bello M.L., Nuccetelli M., Caccuri A.M., Stella L., Parker M.W., Rossjohn J., McKinstry W.J., Mozzi A.F., Federici G., Polizio F., Pedersen J.Z., Ricci G. Human Glutathione Transferase P1-1 and Nitric Oxide Carriers; a New Role for an Old Enzyme. J. Biol. Chem. 2001; 276(45)42138–42145
  • Adler V., Yin Z., Fuchs S.Y., Benezra M., Rosario L., Tew K.D., Pincus M.R., Sardana M., Henderson C.J., Wolf C.R., Davis R.J., Ronai Z. Regulation of JNK Signaling by GSTp. EMBO J. 1999; 18(5)1321–1334
  • Monaco R., Friedman F.K., Hyde M.J., Chen J.M., Manolatus S., Adler V., Ronai Z., Koslosky W., Pincus M.R. Identification of a Glutathione-S-transferase Effector Domain for Inhibition of Jun Kinase, by Molecular Dynamics. J. Protein Chem. 1999; 18(8)859–866
  • Wang T., Arifoglu P., Ronai Z., Tew K.D. Glutathione S-Transferase P1-1 (GSTP1-1) Inhibits c-Jun N-Terminal Kinase (JNK1) Signaling Through Interaction with the C-Terminus. J. Biol. Chem. 2001; 276(24)20999–21003
  • Barr R.K., Bogoyevitch M.A. The c-Jun N-Terminal Kinase Family of Mitogen-Activated Protein Kinases (JNK MAPKs). Int. J. Biochem. Cell Biol. 2001; 33: 1047–1063
  • Bernardini S., Bernassola F., Cortese C., Ballerini S., Melino G., Motti C., Bellincampi L., Iori R., Federici G. Modulation of GST P1-1 Activity by Polymerization During Apoptosis. J. Cell Biochem. 2000; 77(4)645–653
  • Yin Z., Ivanov V.N., Habelhah H., Tew K., Ronai Z. Glutathione S-Transferase p Elicits Protection Against H2O2-Induced Cell Death via Coordinated Regulation of Stress Kinases. Cancer Res. 2000; 60(15)4053–4057
  • Van Haaften R.I., den Hartog G.J., Evelo C.T., Haenen G.R., Bast A. Hypochlorous Acid Is a Potent Inhibitor of Gst P1-1. Chem. Biol. Interact. 2001; 138(1)77–83
  • Villafania A., Anwar K., Amar S., Chie L., Way D., Chung D.L., Adler V., Ronai Z., Brandt-Rauf P.W., Yamaizumii Z., Kung H.F., Pincus M.R. Glutathione-S-transferase as a Selective Inhibitor of Oncogenic Ras-p21-Induced Mitogenic Signaling Through Blockade of Activation of Jun by Jun-N-terminal Kinase. Ann. Clin. Lab. Sci. 2000; 30(1)57–64
  • Cho S.G., Lee Y.H., Park H.S., Ryoo K., Kang K.W., Park J., Eom S.J., Kim M.J., Chang T.S., Choi S.Y., Shim J., Kim Y., Dong M.S., Lee M.J., Kim S.G., Ichijo H., Choi E.J. Glutathione S-Transferase Mu Modulates the Stress-Activated Signals by Suppressing Apoptosis Signal-Regulating Kinase 1. J. Biol. Chem. 2001; 276(16)12749–12755
  • Dulhunty A., Gage P., Curtis S., Chelvanayagam G., Board P. The Glutathione Transferase Structural Family Includes a Nuclear Chloride Channel and a Ryanodine Receptor Calcium Release Channel Modulator. J. Biol. Chem. 2001; 276(5)3319–3323
  • Kampranis S.C., Damianova R., Atallah M., Toby G., Kondi G., Tsichlis P.N., Makris A.M. A Novel Plant Glutathione S-Transferase/Peroxidase Suppresses Bax Lethality in Yeast. J. Biol. Chem. 2000; 275(38)29207–29216
  • Buller A.L., Clapper M.L., Tew K.D. Glutathione S-Transferases in Nitrogen Mustard-Resistant and Sensitive Cell Lines. Mol. Pharmacol. 1987; 31: 575–580
  • Yang W.Z., Begleiter A., Johnston J.B., Israels L.G., Mowat M.R. Role of Glutathione and Glutathione S-Transferases in Chlorambucil Resistance. Mol. Pharmacol. 1992; 41: 625–630
  • Berhane K., Hao X.Y., Egyhåzy S., Hansson J., Ringborg U., Mannervik B. Contribution of Glutathione S-Transferase M3-3 to 1,3-Bis(2-Chloroethyl)-1-nitrosourea Resistance in a Human Non-small Cell Lung Cancer Cell Line. Cancer Res. 1993; 53: 4257–4261
  • Skalski V., Yarosh B.D., Batist G., Gros P., Feindel W., Kopriva D., Panasci L.C. Mechanisms of Resistance to (2-Chloroethyl)-3-sarcosinamide-1-nitrosurea (sarCNU) in Sensitive and Resistant Human Glioma Cells. Mol. Pharmacol. 1990; 38: 299–305
  • Dirven H.A., Megens L., Oudshoorn M.J., Dingemanse M.A., van Ommen B., van Bladeren P.J. Glutathione Conjugation of the Cytostatic Drug Ifosfamide and the Role of Human Glutathione S-Transferases. Chem. Res. Toxicol. 1995; 8: 979–986
  • Dirven H.A., Dictus E.L., Broeders N.L., van Ommen B., van Bladeren P.J. The Role of Human Glutathione S-Transferase Isoenzymes in the Formation of Glutathione Conjugates of the Alkylating Cytostatic Drug Thiotepa. Cancer Res. 1995; 55(8)1701–1706
  • Tsuchida S., Sato K. Glutathione Transferases and Cancer. Crit. Rev. Biochem. Mol. Biol. 1992; 27(4–5)337–384
  • Tew K.D. Glutathione-Associated Enzymes in Anticancer Drug Resistance. Cancer Res. 1994; 54(16)4313–4320
  • Tew K., Monks A., Barone L., Rosser D., Akerman G., Montali J.A., Wheatley J.B., Schmidt D.E. Glutathione-Associated Enzymes in the Human Cell Lines of the National Cancer Institute Drug Screening Program. Mol. Pharmacol. 1996; 50: 149–159
  • Morrow C.S., Cowan K.H. Antineoplastic Drug Resistance and Breast Cancer. Ann. NY Acad. Sci. 1993; 698: 289–312
  • Niitsu Y., Takahashi Y., Ban N., Takayama T., Saito T., Katahira T., Umetsu Y., Nakajima T., Ohi M., Kuga T., Sakamaki S., Matsunaga T., Hirayama Y., Kuroda H., Homma H., Kato J., Kogawa K. A Proof of Glutathione S-Transferase-pi-related Multidrug Resistance by Transfer of Antisense Gene to Cancer Cells and Sense Gene to Bone Marrow Stem Cell. Chem. Biol. Interact. 1998; 111–112: 325–332
  • Nakagawa K., Saijo N., Tsuchida S., Sakai M., Tsunokawa Y., Yokota J., Muramatsu M., Sato K., Terada M., Tew K.D. Glutathione-S-transferase Pi as a Determinant of Drug Resistance in Transfectant Cell Lines. J. Biol. Chem. 1990; 265(8)4296–4301
  • Harbottle A., Daly A.K., Atherton K., Campbell F.C. Role of Glutathione S-Transferase P1, P-Glycoprotein and Multidrug Resistance-Associated Protein 1 in Acquired Doxorubicin Resistance. Int. J. Cancer 2001; 92: 777–783
  • Asakura T., Hashizume Y., Tashiro K., Searashi Y., Ohkawa K., Nishihira J., Sakai M., Shibasaki T. Suppression of GST-P by Treatment with Glutathione–Doxorubicin Conjugate Induces Potent Apoptosis in Rat Hepatoma Cells. Int. J. Cancer 2001; 94(2)171–177
  • Gaudiano G., Koch T.H., LoBello M., Nuccetelli M., Ravagnan G., Serafino A., Sinibaldi-Vallebona P. Lack of Glutathione Conjugation to Adriamycin in Human Breast Cancer MCF-7/DOX Cells. Inhibition of Glutathione S-Transferase P1-1 by Glutathione Conjugates from Anthracyclines. Biochem. Pharmacol. 2000; 60: 1915–1923
  • Sinha B.K., Mimnaugh E.G. Free Radicals and Anticancer Drug Resistance-Oxygen Free Radicals in the Mechanism of Drug Cytotoxicity and Resistance by Certain Tumors. Free Radic. Biol. Med. 1990; 8: 567–581
  • Datta K., Kulkarni A.P. Inhibition of Mammalian Hepatic Glutathione S-Transferases by Acetylenic Fatty Acids. Toxicol. Lett. 1994; 73(2)157–165
  • Mitra A., Govindwar S., Joseph P., Kulkarni A. Inhibition of Human Term Placental and Fetal Liver Glutathione-S-transferases by Fatty Acids and Fatty Acid Esters. Toxicol. Lett. 1992; 60(3)281–288
  • Kulkarni A.A., Kulkarni A.P. Retinoids Inhibit Mammalian Glutathione Transferases. Cancer Lett. 1995; 91(2)185–189
  • Van Haaften R.I., Evelo C.T., Haenen G.R., Bast A. Alpha-Tocopherol Inhibits Human Glutathione S-Transferase Pi. Biochem. Biophys. Res. Commun. 2001; 280(3)631–633
  • Van Haaften R.I., Evelo C.T., Penders J., Eijnwachter M.P., Haenen G.R., Bast A. Inhibition of Human Glutathione S-Transferase P1-1 by Tocopherols and Alpha-Tocopherol Derivatives. Biochim. Biophys. Acta 2001; 1548(1)23–28
  • Listowski I. High Capacity Binding by Glutathione S-Transferases and Glucocorticoid Resistance. Structure and Function of Glutathione Transferases, K.D. Tew, C.B. Pickett, T.J. Mantle, B. Mannervik, J.D. Hayes. CRC Press, Boca Raton 1999; 199–209
  • Ploemen J.H., van Ommen B., van Bladeren P.J. Inhibition of Rat and Human Glutathione S-Transferase Isoenzymes by Ethacrynic Acid and Its Glutathione Conjugate. Biochem. Pharmacol. 1990; 40(7)1631–1635
  • Meyer D.J. Significance of an Unusually Low Km for Glutathione in Glutathione Transferases of the α, μ, and π Classes. Xenobiotica 1993; 23(8)823–834
  • Ouwerkerk-Mahadevan S., van Boom J.H., Dreef T.M., Ploemen J.H., Meyer D.J., Mulder G.J. Glutathione Analogues as Novel Inhibitors of Rat and Human Glutathione S-Transferase Isoenzymes, as Well as of Glutathione Conjugation in Isolated Rat Hepatocytes and in the Rat In Vivo. Biochem. J. 1995; 308(Pt 1)283–290
  • Ouwerkerk-Mahadevan S., Tirona R.G., Ripping R.A., Ploemen J.H., van Bladeren P.J., Pang K.S., van Boom J.H., Mulder G.J. Inhibition of Glutathione Conjugation by Glutathione Analogues in the Perfused Rat Liver. Effect of Esterification on the Potency of Gamma-l-glutamyl-alpha-(d-2-aminoadipyl)-N-2-heptylamine. Drug Metab. Dispos. 1997; 25(10)1137–1143
  • Ouwerkerk-Mahadevan S., van Boom J.H., Mulder G.J. Isoenzyme-Selective Inhibition of Glutathione Conjugation In Vivo: Selective Inhibition of the Conjugation of S-2-Bromoisovalerylurea in the Rat. J. Pharmacol. Exp. Ther. 1996; 276(3)923–928
  • Koehler R.T., Villar H.O., Bauer K.E., Higgins D.L. Ligand-Based Protein Alignment and Isozyme Specificity of Glutathione S-Transferase Inhibitors. Proteins 1997; 28(2)202–216
  • Morgan A.S., Ciaccio P.J., Tew K.D., Kauvar L.M. Isozyme-Specific Glutathione S-Transferase Inhibitors Potentiate Drug Sensitivity in Cultured Human Tumor Cell Lines. Cancer Chemother. Pharmacol. 1996; 37(4)363–370
  • Chen Y.R., Wang X., Templeton D., Davis R.J., Tan T.H. The Role of c-Jun N-Terminal Kinase (JNK) in Apoptosis Induced by Ultraviolet C and Gamma Radiation. Duration of JNK Activation May Determine Cell Death and Proliferation. J. Biol. Chem. 1996; 271(50)31929–31936
  • van der Kolk D.M., Vellenga E., Muller M., de Vries E.G. Multidrug Resistance Protein MRP1, Glutathione, and Related Enzymes. Their Importance in Acute Myeloid Leukemia. Adv. Exp. Med. Biol. 1999; 457: 187–198
  • Satoh K. Weak Electrophile Selective Characteristics of the Rat Preneoplastic Marker Enzyme Glutathione S-Transferase P-Form, GST-P(7-7): A Theory of Linear Free Energy Relationships for Evaluation of the Active Site Hydrophobicity of Isoenzymes. Carcinogenesis 1998; 19(9)1665–1671
  • Satoh K., Sato R., Takahata T., Suzuki S., Hayakari M., Tsuchida S., Hatayama I. Quantitative Differences in the Active-Site Hydrophobicity of Five Human Glutathione S-Transferase Isoenzymes: Water-Soluble Carcinogen-Selective Properties of the Neoplastic GSTP1-1 Species. Arch. Biochem. Biophys. 1999; 361(2)271–276
  • Berhane K., Widersten M., Engstrom A., Kozarich J.W., Mannervik B. Detoxication of Base Propenals and Other Alpha, Beta-Unsaturated Aldehyde Products of Radical Reactions and Lipid Peroxidation by Human Glutathione Transferases. Proc. Natl Acad. Sci. USA 1994; 91(4)1480–1484
  • Smith G.C.M., Jackson S.P. The DNA-Dependent Protein Kinase. Gen. Dev. 1999; 13: 916–934
  • Lees-Miller S.P. The DNA-Dependent Protein Kinase, DNA-PK: 10 Years and No Ends in Sight. Biochem. Cell. Biol. 1996; 74(4)503–512
  • Park S.J., Oh E.J., Yoo M.A., Lee S.H. Involvement of DNA-Dependent Protein Kinase in Regulation of Stress-Induced JNK Activation. DNA Cell. Biol. 2001; 20(10)637–645
  • Hammersten O., Chu G. DNA-Dependent Protein Kinase–DNA Binding and Activation in Absence of Ku. Proc. Natl Acad. Sci. USA 1998; 95: 525–530
  • D'Arpa P., Liu L.F. Topoisomerase Targeting Antitumor Drugs. Biochim. Biophys. Acta 1989; 989: 163–177
  • Shen H., Schultz M., Kruh G.D., Tew K.D. Increased Expression of DNA-Dependent Protein Kinase Confers Resistance to Adriamycin. Biochim. Biophys. Acta 1998; 1381(2)131–138
  • Kim S.H., Um J.H., Dong-Won B., Kwon B.H., Kim D.W., Chung B.S., Kang C.D. Potentiation of Chemosensitivity in Multidrug-Resistant Human Leukemia CEM Cells by Inhibition of DNA-Dependent Protein Kinase Using Wortmannin. Leuk. Res. 2000; 24(11)917–925
  • Martin N.M. DNA Repair Inhibition and Cancer Therapy. J. Photochem. Photobiol. B 2001; 63(1–3)162–170
  • Izzard R.A., Jackson S.P., Smith G.C. Competitive and Noncompetitive Inhibition of the DNA-Dependent Protein Kinase. Cancer Res. 1999; 59(11)2581–2586
  • Rozenzweig K.E., Youmell M.B., Palayoor S.T., Price B.D. Radiosensitization of Human Tumor Cells by the PI 3-Kinase Inhibitors Wortmannin and Ly294002 Correlates with Inhibition of DNA-Dependent Protein Kinase and Prolonged G2-M Delay. Clin. Cancer Res. 1997; 3: 1149–1156
  • Take Y., Kumano M., Hamano Y., Fukatsu H., Teraoka H., Nishimura S., Okuyama A. OK-1035 a Selective Inhibitor of DNA-Dependent Protein Kinase. Biochem. Biophys. Res. Commun. 1995; 215(1)41–47
  • Take Y., Kumano M., Teraoka H., Nishimura S., Okuyama A. DNA-Dependent Protein Kinase Inhibitor (OK-1035) Suppresses p21 Expression in HCT116 Cells Containing Wild-Type p53 Induced by Adriamycin. Biochem. Biophys. Res. Commun. 1996; 221(2)207–212
  • Stockley M., Clegg W., Fontana G., Golding B.T., Martin N., Rigoreau L.J., Smith G.C., Griffin R.J. Synthesis, Crystal Structure Determination, and Biological Properties of the DNA-Dependent Protein Kinase (DNA-PK) Inhibitor 3-Cyano-6-hydrazonomethyl-5-(4-pyridyl)pyrid-[1H]-2-one(OK-1035). Bioorg. Med. Chem. Lett. 2001; 11(21)2837–2841
  • Eriksson A., Yachnin J., Lewensohn R., Nilsson A., Nilsso A. DNA-Dependent Protein Kinase Is Inhibited by Trifluoperazine. Biochem. Biophys. Res. Commun. 2001; 283(4)726–731
  • Shen H., Schultz M.P., Tew K.D. Glutathione Conjugate Interactions with DNA-Dependent Protein Kinase. J. Pharmacol. Exp. Ther. 1999; 290(3)1101–1106
  • Jin S., Kharbanda S., Mayers B., Kufe D., Weaver D.T. Binding of Ku and c-Abl at the Kinase Homology Region of DNA-Dependent Protein Kinase Catalytic Subunit. J. Biol. Chem. 1997; 272: 24763–24766
  • Kumar S., Pandey P., Bharti A., Jin S., Weichselbaum R., Weaver D., Kufe D., Kharbanda S. Regulation of DNA-Dependent Protein Kinase by the Lyn Tyrosine Kinase. J. Biol. Chem. 1998; 273: 25654–25658
  • Thornalley P.J. Advances in Glyoxalase Research. Glyoxalase Expression in Malignancy, Anti-proliferative Effects of Methylglyoxal, Glyoxalase I Inhibitor Diesters and S-d-Lactoylglutathione, and Methylglyoxal-Modified Protein Binding and Endocytosis by the Advanced Glycation Endproduct Receptor. Crit. Rev. Oncol. Hematol. 1995; 20: 99–128
  • Thornalley P.J. Glutathione-Dependent Detoxification of Alpha-Oxoaldehydes by the Glyoxalase System: Involvement in Disease Mechanisms and Antiproliferative Activity of Glyoxalase I Inhibitors. Chem. Biol. Interact. 1998; 111–112: 137–151
  • Larsen K., Aronsson A.C., Marmstal E., Mannervik B. Immunological Comparison of Glyoxalase I from Yeast and Mammals and Quantitative Determination of the Enzyme in Human Tissues by Radioimmunoassay. Comp. Biochem. Physiol. B 1985; 82(4)625–638
  • Mannervik B., Ridderstrom M. Catalytic and Molecular Properties of Glyoxalase I. Biochem. Soc. Trans. 1993; 21(2)515–517
  • Vander Jagt D.L. Glyoxalase II: Molecular Characteristics, Kinetics and Mechanism. Biochem. Soc. Trans. 1993; 21(2)522–527
  • Castro V.M., Siderstrom M., Carlberg I., Widersten M., Platz A., Mannervik B. Differences Among Human Tumor Cell Lines in the Expression of Glutathione Transferase and Other Glutathione-Linked Enzymes. Carcinogenesis 1990; 11: 1569–1576
  • Ayoub F.M., Zaman M.A., Thornalley P.J. Glyoxalase Activity in Human Tumour Cell Lines In Vitro. Biochem. Soc. Trans. 1993; 21: 176S
  • Ranganathan S., Tew K. Analysis of Glyoxalase I from Normal and Tumour Tissue from Human Colon. Biochim. Biophys. Acta 1994; 1170: 17–24
  • Sakamoto H., Mashima T., Kizaki A., Dan S., Hashimoto Y., Naito M., Tsuruo T. Glyoxalase I Is Involved in Resistance of Human Leukemia Cells to Antitumor Induced Apoptosis. Blood 2000; 95: 3214–3218
  • Dills W.L. Nutritional and Physiological Consequences of Tumour Glycolysis. Parasitology 1993; 107: S177–S186
  • Vince R., Wadd W.B. Glyoxalase Inhibitors as Potential Anticancer Agents. Biochem. Biophys. Res. Commun. 1969; 35: 593–598
  • Ayoub F.M., Zaman M.A., Thornalley P.J. Glyoxalase Activities in Human Tumour Cell Lines In Vitro. Anticancer Res. 1993; 13: 151–156
  • Vince R., Daluge S., Wadd W.B. Studies on the Inhibition of Glyoxalase I by S-Substituted Glutathiones. J. Med. Chem. 1971; 14(5)402–404
  • Lo T.W.C., Thornalley P.J. Inhibition of Proliferation of Human Leukemia 60 Cells by Diethyl Esters of Glyoxalase Inhibitors In Vitro. Biochem. Pharmacol. 1992; 44: 2357–2363
  • Conroy P.J. Carcinostatic Activity of Methylglyoxal and Related Substances in Tumour Bearing Mice. Ciba Found. Symp. 1979; 67: 271–298
  • Thornalley P.J., Ladan M.J., Ridgway S.J., Kang Y. Antitumor Activity of S-(p-Bromobenzyl)Glutathione Diesters In Vitro: A Structure–Activity Study. J. Med. Chem. 1996; 39(17)3409–3411
  • Thornalley P.J., Edwards L.G., Kang Y., Wyatt C., Davies N., Ladan M.J., Double J. Antitumour Activity of S-p-Bromobenzylglutathione Cyclopentyl Diester In Vitro and In Vivo. Inhibition of Glyoxalase I and Induction of Apoptosis. Biochem. Pharmacol. 1996; 51(10)1365–1372
  • Doweyko A.M. The Hypothetical Active Site Lattice. An Approach to Modelling Active Sites from Data on Inhibitor Mechanism. J. Med. Chem. 1988; 31: 1396–1406
  • Klopman G., Dimayuga M.L. Computer Automated Structure Evaluation of Flavonoids and Other Structurally Related Compounds as Glyoxalase I Enzyme Inhibitors. Mol. Pharmacol. 1988; 34: 218–222
  • Thomson C., Higgins D., Edge C. Theorectic Studies of the Electrostatic Potential of Some Enzyme Inhibitors Using Computer Graphics Techniques. J. Mol. Graphics 1988; 6: 171–177
  • Thomson C., Brandt R. Theoretical Investigations of the Structure of Potential Inhibitors of the Enzyme Glyoxalase I. Int. J. Quantum Chem. 1983; 10: 357–373
  • Cameron A.D., Olin B., Ridderstrom M., Mannervik B., Jones T.A. Crystal Structure of Human Glyoxalase I—Evidence for Gene Duplication and 3D Domain Swapping. EMBO J. 1997; 16: 3386–3395
  • Johansson A.S., Ridderstrom M., Mannervik B. The Human Glutathione Transferase P1-1 Specific Inhibitor TER 117 Designed for Overcoming Cytostatic-Drug Resistance Is Also a Strong Inhibitor of Glyoxalase I. J. Pharmacol. Exp. Ther. 2000; 57: 619–624
  • Ridderstrom M., Cameron A.D., Jones T.A., Mannervik B. Mutagenesis of Residue 157 in the Active Site of Human Glyoxalase I. Biochem. J. 1997; 328: 231–235
  • Ridderstrom M., Jemth P., Cameron A.D., Mannervik B. The Active-Site Residue Tyr-175 in Human Glyoxalase II Contributes to Binding of Glutathione Derivatives. Biochim. Biophys. Acta 2000; 1481(2)344–348
  • Cameron A.D., Ridderstrom M., Olin B., Kavarana M.J., Creighton D.J., Mannervik B. Reaction Mechanism of Glyoxalase I Explored by an X-Ray Crystallographic Analysis of the Human Enzyme in Complex with a Transition State Analogue. Biochemistry 1999; 38: 13480–13490
  • Hamilton D.S., Creighton D.J. Inhibition of Glyoxalase I by the Enediol Mimic S-(N-Hydroxy-N-methylcarbamoyl)Glutathione. The Possible Basis of a Tumor-Selective Anticancer Strategy. J. Biol. Chem. 1992; 267(35)24933–24936
  • Kalsi A., Kavarana M.J., Lu T., Whalen D.L., Hamilton D.S., Creighton D.J. Role of Hydrophobic Interactions in Binding S-(N-Aryl/Alkyl-N-hydroxycarbamoyl) Glutathiones to the Active Site of the Antitumor Target Enzyme Glyoxalase I. J. Med. Chem. 2000; 43: 3981–3986
  • Sharkey E.M., O'Neill H.B., Kavarana M.J., Wang H., Creighton D.J., Sentz D.L., Eiseman J.L. Pharmacokinetics and Antitumor Properties in Tumor-Bearing Mice of an Enediol Analogue Inhibitor of Glyoxalase I. Cancer Chemother. Pharmacol. 2000; 46(2)156–166
  • Lo T.W., Thornalley P.J. Inhibition of Proliferation of Human Leukaemia 60 Cells by Diethyl Esters of Glyoxalase Inhibitors In Vitro. Biochem. Pharmacol. 1992; 44(12)2357–2363
  • Al Timari A., Douglas K.T. Inhibition by Glutathione Derivatives of Bovine Liver Glyoxalase II (Hydroxyacylglutathione Hydrolase) as a Probe of the N- and S-Sites for Substrate Binding. Biochim. Biophys. Acta 1986; 870(2)219–225
  • Lo T.W., Thornalley P.J. Inhibition of Human Leukaemia 60 Cell Growth by Diethyl Esterification of the Glyoxalase II Inhibitor S-p-Nitrobenzoxycarbonylglutathione In Vitro. Biochem. Soc. Trans. 1993; 21(2)159S
  • Thornalley P.J., Tisdale M.J. Inhibition of Proliferation of Human Promyelocytic Leukemia HL60 Cells by S-d-Lactoyl-glutathione. Leuk. Res. 1988; 12: 897–904
  • Edwards L.G., Clelland J.D., Thornalley P.J. Characteristics of the Inhibition of Human Promyelocytic Leukemia HL60 Cell-Growth by S-D-Lactoylglutathione In Vitro. Leuk. Res. 1993; 17(4)305–310
  • Reeves M.E., Thornalley P.J. The Hydrolysis of S-d-Latoyl-glutathione. Biochem. Soc. Trans. 1993; 21: 169S
  • Christopherson R.I., Schmalzl K.J., Szabados E., Goodridge R.J., Harsanyi M.C., Sant M.E., Algar E.M., Anderson J.E., Armstrong A., Sharma S.C. Mercaptan and Dicarboxylate Inhibitors of Hamster Dihydroorotase. Biochemistry 1989; 28(2)463–470
  • Edwards L.G., Thornalley P.J. Prevention of S-d-Lactoylglutathione-induced Inhibition of Human Leukaemia 60 Cell Growth by Uridine. Leuk. Res. 1994; 18(9)717–722
  • Edwards L.G., Adesida A., Thornalley P.J. Inhibition of Human Leukaemia 60 Cell Growth by S-d-Lactoylglutathione In Vitro. Mediation by Metabolism to N-d-Lactoylcysteine and Induction of Apoptosis. Leuk. Res. 1996; 20(1)17–26
  • Witz G. Biological Interactions of a,b-Unsaturated Aldehydes. J. Free Radic. Biol. Med. 1989; 7: 333–349
  • Ploemen J.H., Van Schanke A., van Ommen B., van Bladeren P.J. Reversible Conjugation of Ethacrynic Acid with Glutathione and Human Glutathione S-Transferase P1-1. Cancer Res. 1994; 54(4)915–919
  • Ishikawa T., Kobayashi K., Sogame Y., Hayashi K. Evidence for Leukotriene C4 Transport Mediated by an ATP-Dependent Glutathione S-Conjugate Carrier in Rat Heart and Liver Plasma Membranes. FEBS Lett. 1989; 29(1)95–98
  • Ishikawa T. The ATP-Dependent Glutathione S-Conjugate Export Pump. Trends Biochem. Sci. 1992; 17(11)463–468
  • Leier I., Jedlitschky G., Buchholz U., Cole S.P., Deeley R.G., Keppler D. The MRP Gene Encodes an ATP Dependent Export Pump for Leukotriene C4 and Structurally Related Conjugates. J. Biol. Chem. 1994; 269(45)27807–27810
  • Martinoia E., Grill E., Tommassini R., Kreuz K., Amrhein N. ATP-Dependent Glutathione S-Conjugate “Export” Pump in Vacuolar Membrane of Plants. Nature 1993; 364: 247–249
  • Muller M., Meijer C., Zaman G.J., Borst P., Scheper R.J., Mulder N.H., de Vries E.G., Jansen P.L. Overexpression of the Gene Encoding the Multidrug Resistance-Associated Protein Results in Increased ATP-Dependent Glutathione S-Conjugate Transport. Proc. Natl Acad. Sci. USA 1994; 91(26)13033–13037
  • Jedlitschky G., Leier I., Buchholz U., Center M., Keppler D. ATP-Dependent Transport of Glutathione S-Conjugates by the Multidrug Resistance-Associated Protein. Cancer Res. 1994; 54(18)4833–4836
  • Ishikawa T., Tien Kuo M., Furuta K., Suzuki M. The Human Multidrug Resistance-Associated Protein (MRP) Gene Family: From Biological Function to Drug Molecular Design. Clin. Chem. Lab. Med. 2000; 38(9)893–897
  • Borst P., Evers R., Kool M., Wijnholds J. A Family of Drug Transporters: The Multidrug Resistance-Associated Proteins. J. Natl Cancer Inst. 2000; 92(16)1295–1302
  • Renes J., de Vries E.G., Jansen P.L., Muller M. The (Patho)Physiological Functions of the MRP Family. Drug Resist. Updat. 2000; 3(5)289–302
  • Leslie E.M., Deeley R.G., Cole S.P. Toxicological Relevance of the Multidrug Resistance Protein 1. MRP1 (ABCC1) and Related Transporters. Toxicology 2001; 167(1)3–23
  • Rappa G., Lorico A., Flavell R.A., Sartorelli A.C. Evidence That the Multidrug Resistance Protein (MRP) Functions as a Co-transporter of Glutathione and Natural Product Toxins. Cancer Res. 1997; 57(23)5232–5237
  • Loe D.W., Deeley R.G., Cole S.P. Characterization of Vincristine Transport by the M(r) 190,000 Multidrug Resistance Protein (MRP): Evidence for Cotransport with Reduced Glutathione. Cancer Res. 1998; 58(22)5130–5136
  • Zaman G.J., Lankelma J., van Tellingen O., Beijnen J., Dekker H., Paulusma C., Oude Elferink R.P., Baas F., Borst P. Role of Glutathione in the Export of Compounds from Cells by the Multidrug-Resistance-Associated Protein. Proc. Natl Acad. Sci. USA 1995; 92(17)7690–7694
  • Paulusma C.C., van-Geer M.A., Evers R., Heijn M., Ottenhoff R., Borst P., Oude E.R. Canalicular Multispecific Organic Anion Transporter/Multidrug Resistance Protein 2 Mediates Low-Affinity Transport of Reduced Glutathione. Biochem. J. 1999; 338(Pt 2)393–401
  • Leier I., Jedlitschky G., Buchholz U., Center M., Cole S.P., Deeley R.G., Keppler D. ATP-Dependent Glutathione Disulphide Transport Mediated by the MRP Gene-Encoded Conjugate Export Pump. Biochem. J. 1996; 314(Pt 2)433–437
  • Heijn M., Hooijberg J.H., Scheffer G.L., Szabo G., Westerhoff H.V., Lankelma J. Anthracyclines Modulate Multidrug Resistance Protein (MRP) Mediated Organic Anion Transport. Biochim. Biophys. Acta 1997; 1326(1)12–22
  • König J., Nies A.T., Cui Y., Leier I., Keppler D. Conjugate Export Pumps of the Multidrug Resistance Protein (MRP) Family: Localization, Substrate Specificity, and MRP2-Mediated Drug Resistance. Biochim. Biophys. Acta 1999; 1461(2)377–394
  • Loe D.W., Deeley R.G., Cole S.P.C. Biology of the Multidrug Resistance-Associated Protein, MRP. Eur. J. Cancer 2002; 32A: 945–957
  • Flens M.J., Zaman G.J., van der Valk P., Izquierdo M.A., Schroeijers A.B., Scheffer G.L., de Haas M., Meijer C.J.L.M., Scheper R.J. Tissue Distribution of the Multidrug Resistance Associated Protein. Am. J. Pathol. 1996; 148: 1237–1247
  • Deeley R.G., Cole S.P.C. Function, Evolution and Structure of the Multidrug Resistance Protein (MRP). Semin. Cancer Biol. 1997; 8: 193–204
  • Keppler D., König J. Expression and Localization of the Conjugate Export Pump Encoded by the MRP2 (cMRP/CMOAT) Gene in Liver. FASEB J. 1997; 11: 509–516
  • Schaub T.P., Kartenbeck J., Konig J., Vogel O., Witzgall R., Kriz W., Keppler D. Expression of the Conjugate Export Pump Encoded by the MRP2 Gene in the Apical Membrane of Kidney Proximal Tubules. J. Am. Soc. Nephrol. 1997; 8: 1213–1221
  • Jansen P.L.M., Peters W.H., Lamers W.H. Hereditary Chronic Conjugated Hyperbilirubinemia in Mutant Rats Caused by Defective Hepatic Anion Transport. Hepatology 1985; 5: 573–579
  • Mikami T., Nozaki T., Tagaya O. The Characteristics of a New Mutant in Rats with Hyperbilirubinurea Syndrome. Congenital Anom. 1986; 26: 250–251
  • Paulusma C.C., Kool M., Bosma P.J., Scheffer G.L., Ter Borg F., Scheper R.J., Tytgat G.N., Borst P., Baas F., Oude Elferink R.P. A Mutation in the Human Canalicular Multispecific Organic Anion Transporter Gene Causes the Dubin-Johnson Syndrome. Hepatology 1997; 25: 1539–1542
  • Gottesman M.M., Pastan I. Biochemistry of Multidrug Resistance Mediated by the Multidrug Transporter. Annu. Rev. Biochem. 1993; 62: 385–427
  • McGrath T., Center M.S. Adriamycin Resistance in HL60 Cells in the Absence of Detectable P-Glycoprotein. Biochem. Biophys. Res. Commun. 1987; 145: 1171–1176
  • Mirski S.E., Gerlach J.H., Cole S.P.C. Multidrug Resistance in a Human Small Cell Lung Cancer Cell Line Selected in Adriamycin. Cancer Res. 1987; 47: 2594–2598
  • Cole S.P.C., Bhardwaj G., Gerlach J.H. Overexpression of a Transporter Gene in a Multidrug-Resistant Human Lung Cancer Cell Line. Science 1992; 258: 1650–1654
  • Hipfner D.R., Deeley R.G., Cole S.P. Structural, Mechanistic and Clinical Aspects of MRP1. Biochim. Biophys. Acta 1999; 1461(2)359–376
  • Litman T., Druley T.E., Stein W.D., Bates S.E. From MDR to MXR: New Understanding of Multidrug Resistance Systems, Their Properties and Clinical Significance. Cell. Mol. Life Sci. 2001; 58(7)931–959
  • Ota E., Abe Y., Oshika Y., Ozeki Y., Iwasaki M., Inoue H., Yamazaki H., Ueyama Y., Takagi K., Ogata T., Tamaoki N., Nakamura M. Expression of the Multidrug Resistance-Associated Protein (MRP) Gene in Non-small Cell Lung Cancer. Br. J. Cancer 1995; 72: 550–554
  • Nooter K., Brutel De La Riviere G., Klijn J., Stoter G., Foekens J. Multidrug Resistance Protein in Recurrent Breast Cancer. Lancet 1997; 349: 1885–1886
  • Filipits M., Stranzl T., Pohl G., Suchomel R.W., Zochbauer S., Brunner R. MRP Expression in Acute Myeloid Leukemia, an Update. Adv. Exp. Med. Biol. 1999; 457: 141–150
  • Arts H.J., Katsaros D., de Vries E.G., Massobrio M., Genta F., Danese S., Arisio R., Scheper R.J., Kool M., Scheffer G.L., Willemse P.H., van der Zee A.G., Suurmeijer A.J. Drug Resistance-Associated Markers P-Glycoprotein, Multidrug Resistance-Associated Protein 1, Multidrug Resistance-Associated Protein 2, and Lung Resistance Protein as Prognostic Factors in Ovarian Carcinoma. Clin. Cancer Res. 1999; 5(10)2798–2805
  • Yokoyama Y., Sato S., Fukushi Y., Sakamoto T., Futagami M., Saito Y. Significance of Multi-drug-resistant Proteins in Predicting Chemotherapy Response and Prognosis in Epithelial Ovarian Cancer. J. Obstet. Gynaecol. Res. 1999; 25(6)387–394
  • Lorico A., Nesland J., Emilsen E., Fodstad O., Rappa G. Role of the Multidrug Resistance Protein 1 Gene in the Carcinogenicity of Aflatoxin B1: Investigations Using MRP1-Null Mice. Toxicology 2002; 171(2–3)201–205
  • Koike K., Kawabe T., Tanaka T., Toh S., Uchiumi T., Wada M. A Canalicular Multispecific Organic Anion Transporter (cMOAT) Antisense cDNA Enhances Drug Sensitivity in Human Hepatic Cancer Cells. Cancer Res. 1997; 57: 5475–5479
  • Cui Y., Konig J., Buchholz J.K., Spring H., Leier I., Keppler D. Drug Resistance and ATP Dependent Conjugate Transport Mediated by the Apical Multidrug Resistance Protein. MRP2, Permanently Expressed in Human and Canine Cells. Mol. Pharmacol. 1999; 55: 929–937
  • Hooijberg J.H., Broxterman H.J., Kool M., Assaraf Y.G., Peters G.J., Noordhuis P., Scheper R.J., Borst P., Pinedo H.M., Jansen G. Antifolate Resistance Mediated by the Multidrug Resistance Proteins MRP1 and MRP2. Cancer Res. 1999; 59: 2532–2535
  • Schaub T.P., Kartenbeck J., König J., Spring H., Dörsam J., Staeler G., Störkel S., Thon W.F., Keppler D. Expression of the MRP2 Gene-Encoded Conjugate Export Pump in Human Kidney Proximal Tubules and in Renal Cell Carcinoma. J. Am. Soc. Nephrol. 1999; 10: 1159–1169
  • Kim H.S., Min Y.D., Choi C.H. Double-Edged Sword of Chemosensitizer: Increase of Multidrug Resistance Protein (MRP) in Leukemic Cells by an MRP Inhibitor Probenecid. Biochem. Biophys. Res. Commun. 2001; 283(1)64–71
  • Ford J.M., Yang J.M., Hait W.N. Effect of Buthionine Sulfoximine on Toxicity of Verapamil and Doxorubicin to Multidrug Resistant Cells and to Mice. Cancer Res. 1991; 51: 67–72
  • O'Dwyer P.J., Hamilton T.C., LaCreta F.P., Gallo J.M., Kilpatrick D., Halbherr T., Brennan J., Bookman M.A., Hoffman J., Young R.C., Comis R.L., Ozols R.F. Phase I Trial of Buthionine Sulfoximine in Combination with Melphalan in Patients with Cancer. J. Clin. Oncol. 1996; 14(1)249–256
  • Bailey H.H., Mulcahy R.T., Tutsch K.D., Arzoomanian R.Z., Alberti D., Tombes M.B., Wilding G., Pomplun M., Spriggs D.R. Phase I. Clinical Trial of Intravenous l-Buthionine Sulfoximine and Melphalan: An Attempt at Modulation of Glutathione. J. Clin. Oncol. 1994; 12(1)194–205
  • Leslie E.M., Mao Q., Oleschuk C.J., Deeley R.G., Cole S.P. Modulation of Multidrug Resistance Protein 1 (MRP1/ABCC1) Transport and Atpase Activities by Interaction with Dietary Flavonoids. Mol. Pharmacol. 2001; 59(5)1171–1180
  • Hooijberg J.H., Broxterman H.J., Heijn M., Fles D.L.A., Lankelma J., Pinedo H.M. Modulation by (Iso)Flavonoids of the Atpase Activity of the Multidrug Resistance Protein. FEBS Lett. 1997; 413: 344–348
  • Hooijberg J.H., Broxterman H.J., Scheffer G.L., Vrasdonk C., Heijn M., de Jong M.C., Scheper R.J., Lankelma J., Pinedo H.M. Potent Interaction of Flavopiridol with MRP1. Br. J. Cancer 1999; 81(2)269–276
  • Walgren R.A., Karnaky K.J., Jr., Lindenmayer G.E., Walle T. Efflux of Dietary Flavonoid Quercetin 4′-Beta-Glucoside Across Human Intestinal Caco-2 Cell Monolayers by Apical Multidrug Resistance-Associated Protein-2. J. Pharmacol. Exp. Ther. 2000; 294(3)830–836
  • Walle U.K., Galijatovic A., Walle T. Transport of the Flavonoid Chrysin and Its Conjugated Metabolites by the Human Intestinal Cell Line Caco-2. Biochem. Pharmacol. 1999; 58(3)431–438
  • Hollo Z., Homolya L., Hegedus T., Sarkadi B. Transport Properties of the Multidrug Resistance-Associated Protein (MRP) in Human Tumour Cells. FEBS Lett. 1996; 383(1–2)99–104
  • Payen L., Courtois A., Campion J.P., Guillouzo A., Fardel O. Characterization and Inhibition by a Wide Range of Xenobiotics of Organic Anion Excretion by Primary Human Hepatocytes. Biochem. Pharmacol. 2000; 60: 1967–1975
  • Gollapudi S., Thadepalli F., Kim C.H., Gupta S. Difloxacin Reverses Multidrug Resistance in HL-60/AR Cells That Overexpress the Multidrug Resistance-Related Protein (MRP) Gene. Oncol. Res. 1995; 7(5)213–225
  • Courtois A., Payen L., Vernhet L., de Vries E.G., Guillouzo A., Fardel O. Inhibition of Multidrug Resistance-Associated Protein (MRP) Activity by Rifampicin in Human Multidrug-Resistant Lung Tumor Cells. Cancer Lett. 1999; 139: 97–104
  • Terashi K., Oka M., Soda H., Fukuda M., Kawabata S., Nakatomi K., Shiozawa K., Nakamura T., Tsukamoto K., Noguchi Y., Suenaga M., Tei C., Kohno S. Interactions of Ofloxacin and Erythromycin with the Multidrug Resistance Protein (MRP) in MRP-Overexpressing Human Leukemia Cells. Antimicrob. Agents Chemother. 2000; 44(6)1697–1700
  • Klokouzas A., Barrand M.A., Hladky S.B. Effects of Clotrimazole on Transport Mediated by Multidrug Resistance Associated Protein 1 (MRP1) in Human Erythrocytes and Tumour Cells. Eur. J. Biochem. 2001; 268(24)6569–6577
  • Gekeler V., Ise W., Sanders K.H., Ulrich W.R., Beck J. The Leukotriene LTD4 Receptor Antagonist MK571 Specifically Modulates MRP Associated Multidrug Resistance. Biochem. Biophys. Res. Commun. 1995; 208(1)345–352
  • Nakano R., Oka M., Nakamura T., Fukuda M., Kawabata S., Terashi K., Tsukamoto K., Noguchi Y., Soda H., Kohno S. A Leukotriene Receptor Antagonist, ONO-(1078) Modulates Drug Sensitivity and Leukotriene C4 Efflux in Lung Cancer Cells Expressing Multidrug Resistance Protein. Biochem. Biophys. Res. Commun. 1998; 251(1)307–312
  • Evers R., Kool M., Smith A.J., van Deemter L., de Haas M., Borst P. Inhibitory Effect of the Reversal Agents V-104, GF120918 and Pluronic L61 on MDR1 Pgp-, MRP1- and MRP2-Mediated Transport. Br. J. Cancer 2000; 83(3)366–374
  • Miller D.W., Batrakova E.V., Kabanov A.V. Inhibition of Multidrug Resistance-Associated Protein (MRP) Functional Activity with Pluronic Block Copolymers. Pharm. Res. 1999; 16(3)396–401
  • Payen L., Delugin L., Courtois A., Trinquart Y., Guillouzo A., Fardel O. The Sulphonylurea Glibenclamide Inhibits Multidrug Resistance Protein (MRP1) Activity in Human Lung Cancer Cells. Br. J. Pharmacol. 2001; 132: 778–784
  • Ren X.Q., Furukawa T., Aoki S., Nakajima T., Sumizawa T., Haraguchi M., Chen Z.S., Kobayashi M., Akiyama S. Glutathione-Dependent Binding of a Photoaffinity Analog of Agosterol A to the C-Terminal Half of Human Multidrug Resistance Protein. J. Biol. Chem. 2001; 276(25)23197–23206
  • Chen Z.S., Aoki S., Komatsu M., Ueda K., Sumizawa T., Furukawa T., Okumura H., Ren X.Q., Belinsky M.G., Lee K., Kruh G.D., Kobayashi M., Akiyama S. Reversal of Drug Resistance Mediated by Multidrug Resistance Protein (MRP) 1 by Dual Effects of Agosterol A on MRP1 Function. Int. J. Cancer 2001; 93(1)107–113
  • Chen Z.S., Furukawa T., Sumizawa T., Ono K., Ueda K., Seto K., Akiyama S.I. ATP-Dependent Efflux of CPT-11 and SN-38 by the Multidrug Resistance Protein (MRP) and Its Inhibition by PAK-104P. Mol. Pharmacol. 1999; 55(5)921–928
  • Chuman Y., Chen Z.S., Seto K., Sumizawa T., Furukawa T., Tani A., Haraguchi M., Niwa K., Yamada K., Aikou T., Akiyama S. Reversal of MRP-Mediated Vincristine Resistance in KB Cells by Buthionine Sulfoximine in Combination with PAK-104P. Cancer Lett. 1998; 129(1)69–76
  • Sumizawa T., Chen Z.S., Chuman Y., Seto K., Furukawa T., Haraguchi M., Tani A., Shudo N., Akiyama S.I. Reversal of Multidrug Resistance-Associated Protein-Mediated Drug Resistance by the Pyridine Analog PAK-104P. Mol. Pharmacol. 1997; 51(3)399–405
  • Norman B.H., Dantzig A.H., Kroin J.S., Law K.L., Tabas L.B., Shepard R.L., Palkowitz A.D., Hauser K.L., Winter M.A., Sluka J.P., Starling J.J. Reversal of Resistance in Multidrug Resistance Protein (MRP1)-Overexpressing Cells by LY329146. Bioorg. Med. Chem. Lett. 1999; 9(23)3381–3386
  • Seelig A., Blatter X.L., Wohnsland F. Substrate Recognition by P-Glycoprotein and the Multidrug Resistance-Associated Protein MRP1: A Comparison. Int. J. Clin. Pharmacol. Ther. 2000; 38(3)111–121
  • Ishikawa T. ATP/Mg2+-Dependent Cardiac Transport System for Glutathione S-Conjugates. A Study Using Rat Heart Sarcolemma Vesicles. J. Biol. Chem. 1989; 264(29)17343–17348
  • Ito K., Olsen S.L., Qiu W., Deeley R.G., Cole S.P. Mutation of a Single Conserved Tryptophan in Multidrug Resistance Protein 1 (MRP1/ABCC1) Results in Loss of Drug Resistance and Selective Loss of Organic Anion Transport. J. Biol. Chem. 2001; 276(19)15616–15624
  • Furuta K., Tomokiyo K., Tien Kuo M., Ishikawa T., Suzuki M. Molecular Design of Glutathione-Derived Biochemical Probes Targeting the GS-X Pump. Tetrahedron 1999; 55: 7529–7540
  • Leslie E.M., Ito K., Upadhyaya P., Hecht S.S., Deeley R.G., Cole S.P.C. Transport of the b-O-Glucuronide Conjugate of the Tobacco-Specific Carcinogen 4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL) by the Multidrug Resistance Protein 1 (MRP1). J. Biol. Chem. 2001; 276(30)27846–27854
  • O'Brien M.L., Vulevic B., Freer S., Boyd J., Shen H., Tew K.D. Glutathione Peptidomimetic Drug Modulator of Multidrug Resistance-Associated Protein. J. Pharmacol. Exp. Ther. 1999; 291(3)1348–1355
  • Meister A., Tate S.S., Ross L.L. Membrane-Bound γ-Glutamyl Transpeptidase. The Enzymes of Biological Membranes, A. Martonosi, 1990; 315–347
  • Hanigan M.H. Gamma-Glutamyl Transpeptidase, a Glutathionase: Its Expression and Function in Carcinogenesis. Chem. Biol. Interact. 1998; 111–112: 333–342
  • McIntyre T.M., Curthoys N.P. Renal Catabolism of Glutathione—Characterization of a Particulate Rat Renal Dipeptidase That Catalyzes the Hydrolysis of Cysteinylglycine. J. Biol. Chem. 1982; 257: 11915–11921
  • Commandeur J.N., Stijntjes G.J., Vermeulen N.P. Enzymes and Transport Systems Involved in the Formation and Disposition of Glutathione S-Conjugates. Role in Bioactivation and Detoxication Mechanisms of Xenobiotics. Pharmacol. Rev. 1995; 47(2)271–330
  • Hanigan M.H., Frierson H.F. Immunohistochemical Detection of γ-Glutamyl Transpeptidase in Normal Human Tissues. J. Histochem. Cytochem. 1996; 44: 1101–1108
  • Lieberman M.W., Wiseman A.L., Shi Z.Z., Carter B.Z., Barrios R.J., Ou Z.N., Chevez-Barrios P., Wand Y., Habib G.M., Goodman J.C., Huang S.L., Lebovitz R.M., Matzuk M.M. Growth Retardation and Cysteine Deficiency in γ-Glutamyl Transpeptidase-Deficient Mice. Proc. Natl Acad. Sci. USA 1996; 93: 7923–7926
  • Will Y., Fischer K.A., Horton R.A., Kaetzel R.S., Brown M.K., Hedstrom O., Lieberman M.W., Reed D.J. Gamma-Glutamyltranspeptidase-Deficient Knockout Mice as a Model to Study the Relationship Between Glutathione Status, Mitochondrial Function, and Cellular Function. Hepatology 2000; 32(4 Pt 1)740–749
  • Meister A. Mitochondrial Changes Associated with Glutathione Deficiency. Biochim. Biophys. Acta 1995; 1271: 35–42
  • Chen X., Carystinos G.D., Batist G. Potential for Selective Modulation of Glutathione in Cancer Chemotherapy. Chem. Biol. Interact. 1998; 111–112: 263–275
  • Schroder C.P., Godwin A.K., O'Dwyer P.J., Tew K., Hamilton T.C., Ozols R.F. Glutathione and Drug Resistance. Cancer Investig. 1996; 14(2)158–168
  • Komlosh A., Volohonsky G., Porat N., Tuby C., Bluvshtein E., Steinberg P., Oesch F., Stark A.A. γ-Glutamyl Transpeptidase and Glutathione Biosynthesis in Non-tumorigenic and Tumorigenic Rat Liver Oval Cell Lines. Carcinogenesis 2001; 22(12)2009–2016
  • Hanigan M.H. Expression of Gamma-Glutamyl Transpeptidase Provides Tumors with a Selective Growth Advantage of Physiologic Concentrations of Cyst(e)ine. Carcinogenesis 1995; 16: 181–185
  • Selvaraj P., Balasubramanian K.A., Hill P.G. Isolation of Gamma-Glutamyl Transpeptidase from Human Primary Hepatoma and Comparison of Its Kinetic and Catalytic Properties with the Enzyme from Normal Adult and Fetal Liver. Enzyme 1981; 26(2)57–63
  • Tennenbaum T., Weiner A.K., Belanger A.J., Glick A.B., Hennings H., Yuspa S.H. The Suprabasal Expression of Alpha 6 Beta 4 Integrin Is Associated with a High Risk for Malignant Progression in Mouse Skin Carcinogenesis. Cancer Res. 1993; 53(20)4803–4810
  • Hanigan M.H., Frierson H.F., Swanson P.E., deYoung B.R. Altered Expression of Gamma-Glutamyl Transpeptidase in Human Tumors. Hum. Pathol. 1999; 30: 300–305
  • deYoung B.R., Frierson H.F., Swanson P.E., Hanigan M.H. γ-Glutamyl Transpeptidase Immunoreactivity in Human Neoplasia. Lab. Invest. 1996; 74, meeting abstract 840
  • Durham J.R., Frierson H.F., Hanigan M.H. Glutamyl Transpeptidase Immunoreactivity in Benign and Malignant Breast Tissue. Breast Cancer Res. Treat. 1997; 45: 55–62
  • Lewis A.L., Hayes J.D., Wolf C.R. Glutathione and Glutathione Dependent Enzymes in Ovarian Adenocarcinoma Cell Lines Derived from a Patient Before and After the Onset of Drug Resistance: Intrinsic Differences and Cell Cycle Effects. Carcinogenesis 1988; 9: 1283–1287
  • Godwin A.K., Meister A., O'Dwyer P.J., Huang C.S., Hamilton T.C., Anderson M.E. High Resistance to Cisplatin in Human Ovarian Cancer Cell Lines Is Associated with Marked Increase of Glutathione Synthesis. Proc. Natl Acad. Sci. USA 1992; 89: 3070–3074
  • Lau H.M., Lewis A.D., Ehsan M.N., Sikic B.I. Multifactorial Mechanisms Associated with Broad Cross-resistance of Ovarian Cancer Cells Selected by Cyanomorpholino Doxorubicin. Cancer Res. 1991; 51: 5181–5187
  • Ikeda Y., Fujii J., Anderson M.E., Tanaguchi N., Meister A. Involvement of Ser-451 and Ser-452 in the Catalysis of Human γ-Glutamyl Transpeptidase. J. Biol. Chem. 1995; 270: 22223–22228
  • Inoue M., Hiratake J., Suzuki H., Kumagai H., Sakaa L. Identification of Catalytic Nucleophile of E. Coli γ-Glutamyl Transpeptidase by γ-Monofluorophosphono Derivative of Glutamic Acid-N-terminal Thr-391 in Small Subunit Is the Nucleophile. Biochemistry 2000; 39: 7764–7771
  • Neil G.L., Berger A.E., McPartland R.P., Grindley G.B., Bloch A. Biochemical and Pharmacological Effects of the Fermentation-Derived Antitumor Agent, (AlphaS5S)-Alpha-amino-3-chloro-4,5-dihydro-5-isoxazoleacetic Acid (AT-125). Cancer Res. 1979; 39: 852–856
  • Gardell S.J., Tate S.S. Affinity Labeling of Gamma-Glutamyl Transpeptidase by Glutamine Antagonists. Effects of the Gamma-Glutamyl Transferase and Proteinase Activities. FEBS Lett. 1980; 122(2)171–174
  • Capraro M.A., Hughey R.P. Use of Acivicin in the Determination of Rate Constants for Turnover of Rat Renal Gamma-Glutamyltranspeptidase. J. Biol. Chem. 1985; 260(6)3408–3412
  • Elfarra A.A., Anders M.W. Renal Processing of Glutathione Conjugates. Role in Nephrotoxicity. Biochem. Pharmacol. 1984; 33(23)3729–3732
  • Monks T.J., Lau S.S., Highet R.J., Gillette J.R. Glutathione Conjugates of 2-Bromohydroquinone Are Nephrotoxic. Drug Metab. Dispos. 1985; 13(5)553–559
  • Hochwald S.N., Harrison L.E., Rose D.M., Anderson M.E., Burt M.E. γ-Glutamyl Transpeptidase Mediation of Tumor Glutathione Utilization In Vivo. J. Natl Cancer Inst. 1996; 88(3/4)193–197
  • Tate S.S., Meister A. Affinity Labeling of Gamma-Glutamyl Transpeptidase and Location of the Gamma-Glutamyl Binding Site on the Light Subunit. Proc. Natl Acad. Sci. USA 1977; 74(3)931–935
  • Tate S.S., Ross M.E. Human Kidney Gamma-Glutamyl Transpeptidase. Catalytic Properties, Subunit Structure, and Localization of the Gamma-Glutamyl Binding Site on the Light Subunit. J. Biol. Chem. 1977; 252(17)6042–6045
  • Stein R.L., DeCicco C., Nelson D., Thomas B. Slow Binding Inhibition of γ-Glutamyl Transpeptidase by γ-Boroglu. Biochemistry 2001; 40: 5804–5811
  • London R.E., Gabel S.A. Development and Evaluation of a Boronate Inhibitor of Gamma-Glutamyl Transpeptidase. Arch. Biochem. Biophys. 2001; 385(2)250–258
  • Aberkane H., Frank P., Galteau M.M., Wellman M. Acivicin Induces Apoptosis Independently of Gamma-Glutamyltranspeptidase Activity. Biochem. Biophys. Res. Commun. 2001; 285(5)1162–1167
  • Lyons S.D., Sant M.E., Christopherson R.I. Cytotoxic Mechanisms of Glutamine Antagonists in Mouse L1210 Leukemia. J. Biol. Chem. 1990; 265(19)11377–11381
  • Bluvshtein E., Glass G.A., Volohonsky G., Yaakubowitz M., Harness E., Smorodinsky N.I., Seidel A., Frank H., Steinberg P., Stark A.A. Inhibition of the Hydrolytic and Transpeptidase Activities of Rat γ-Glutamyl Transpeptidase by Specific Monoclonal Antibodies. Eur. J. Biochem. 1999; 260: 844–854
  • Chang M., Zhang F., Shen L., Pauss N., Alam I., van Breemen R.B., Blond S.Y., Bolton J.L. Inhibition of Glutathione S-Transferase Activity by the Quinoid Metabolites of Equine Estrogens. Chem. Res. Toxicol. 1998; 11: 758–765
  • Chang M., Shin Y.G., van Breemen R.B., Blond S.Y., Bolton J.L. Structural and Functional Consequences of Inactivation of Human Glutathione S-Transferase P1-1 Mediated by the Catechol Metabolite of Equine Estrogens, 4-Hydroxyequilerin. Biochemistry 2001; 40: 4811–4820

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.