Chem. Res. Toxicol. 2008, 21, 2005–2016
2005
Human Arylamine N-Acetyltransferase 1: In Vitro and Intracellular Inactivation by Nitrosoarene Metabolites of Toxic and Carcinogenic Arylamines Li Liu, Carston R. Wagner, and Patrick E. Hanna* Department of Medicinal Chemistry, UniVersity of Minnesota, 308 HarVard Street Southeast, Minneapolis, Minnesota 55455 ReceiVed June 13, 2008
Arylamines (ArNH2) are common environmental contaminants, some of which are confirmed risk factors for cancer. Biotransformation of the amino group of arylamines involves competing pathways of oxidation and N-acetylation. Nitrosoarenes, which are products of the oxidation pathway, are electrophiles that react with cellular thiols to form sulfinamide adducts. The arylamine N-acetyltransferases, NAT1 and NAT2, catalyze N-acetylation of arylamines and play central roles in their detoxification. We hypothesized that 4-nitrosobiphenyl (4-NO-BP) and 2-nitrosofluorene (2-NO-F), which are nitroso metabolites of arylamines that are readily N-acetylated by NAT1, would be potent inactivators of NAT1 and that nitrosobenzene (NO-B) and 2-nitrosotoluene (2-NO-T), which are nitroso metabolites of arylamines that are less readily acetylated by NAT1, would be less effective inactivators. The second order rate constants for inactivation of NAT1 by 4-NO-BP and 2-NO-F were 59200 and 34500 M-1 s-1, respectively; the values for NO-B and 2-NO-T were 25 and 23 M-1 s-1. Densitometry quantification and comparisons of specific activities with those of homogeneous recombinant NAT1 showed that NAT1 constitutes approximately 0.002% of cytosolic protein in HeLa cells. Treatment of HeLa cells with 4-NO-BP (2.5 µM) for 1 h caused a 40% reduction in NAT1 activity, and 4-NO-BP (10 µM) caused a 50% loss of NAT1 activity within 30 min without affecting either glyceraldehyde 3-phosphate dehydrogenase (GAPDH) or glutathione reductase (GR) activities. 2-NO-F (1 µM) inhibited HeLa cell NAT1 activity by 36% in 1 h, and a 10 µM concentration of 2-NO-F reduced NAT1 activity by 70% in 30 min without inhibiting GAPDH or GR. Mass spectrometric analysis of NAT1 from HeLa cells in which NAT1 was overexpressed showed that treatment of the cells with 4-NO-BP resulted in sulfinamide adduct formation. These results indicated that exposure to low concentrations of nitrosoarenes may lead to a loss of NAT1 activity, thereby compromising a critical detoxification process. Introduction The pervasiveness of human exposure to environmental arylamines (aromatic amines) is well-established (1-3). Concern about the impact of arylamine exposure on human health arises from the vast amount of epidemiological and laboratory data on the toxicity, mutagenicity, and carcinogenicity of various members of this class of agents (4-8). The contribution of environmental arylamine exposure to bladder cancer is supported by the findings that bladder cancer patients exhibit elevated levels of arylamine-hemoglobin adducts and that 4-aminobiphenyl (4-ABP)1-DNA adducts are found in bladder tissue obtained from bladder cancer patients (9-12). 4-ABP, a primary arylamine, is a human bladder carcinogen that is present in tobacco smoke, along with monocyclic arylamines for which * To whom correspondence should be addressed. Tel: 612-625-4152. Fax: 612-6124-0139. E-mail:
[email protected]. 1 Abbreviations: AcCoA, acetyl coenzyme A; 4-ABP, 4-aminobiphenyl; DMEM, Dulbecco’s modified Eagle’s medium; DMSO, dimethylsulfoxide; EDTA, ethylenediaminetetraacetic acid; ESI, electrospray ionization; FBS, fetal bovine serum; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; GR, glutathione reductase; GSH, glutathione; GSSG, oxidized glutathione; MALDI, matrix-assisted laser desorption ionization; NAC, N-acetyl-Lcysteine; NAT, arylamine N-acetyltransferase; NO-B, nitrosobenzene; 4-NOBP, 4-nitrosobiphenyl; 2-NO-F, 2-nitrosofluorene; N-OH-2-AAF, N-hydroxy2-acetylaminofluorene; 2-NO-T, 2-nitrosotoluene; PABA, p-aminobenzoic acid; PAS, p-aminosalicylic acid; PBS, phosphate-buffered saline; PMSF, phenylmethanesulfonyl fluoride; PNPA, p-nitrophenyl acetate; Q-TOF, quadrupole-time-of-flight; SMZ, sulfamethazine; TBS, tris-buffered saline.
Figure 1. Metabolism of arylamines and arylamine metabolites.
exposurehasbeenassociatedwithbladdercancerrisk(4,10,12-14). Although smoking of tobacco is a prominent source of arylamine exposure and is a risk factor for bladder cancer, several studies indicate a substantial contribution to exposure from environmental sources other than tobacco smoke (2, 10, 15-17). Metabolic N-hydroxylation is an essential step in the bioactivation of most mutagenic and carcinogenic arylamines (Figure 1a) (18). The resulting N-arylhydroxylamines may undergo conjugation by conversion to N-acetoxy or N-sulfonyloxy esters (Figure 1b), which are electrophilic metabolites that are involved in the initiation of carcinogenesis by forming covalent adducts with DNA (19). Additionally, N-arylhydroxylamines can undergo oxidation to form nitrosoarenes (Figure 1c). Arylnitroso compounds are electrophilic and react readily with nucleophilic thiols, such as those present in proteins and glutathione (GSH) (20-23). Adduct formation between nitrosoarenes and a cysteine sulthydryl of hemoglobin has been used to an advantage for
10.1021/tx800215h CCC: $40.75 2008 American Chemical Society Published on Web 08/30/2008
2006
Chem. Res. Toxicol., Vol. 21, No. 10, 2008
Liu et al.
Experimental Procedures
Figure 2. Structures of nitrosoarenes used in this study.
the quantification of human exposure to environmental arylamines (2, 9, 10, 16, 17, 24). In contrast to the metabolic conversion of an arylamine to an N-hydroxylated metabolite, N-acetylation is considered to be a detoxification reaction (Figure 1d) (7). Humans express two cytosolic, polymorphic N-acetyltransferases, NAT1 and NAT2, which catalyze the N-acetylation of primary arylamines (25, 26). The polymorphisms of NAT2 that cause a slow acetylation phenotype are associated with risk of arylamineinduced bladder cancer and influence the therapeutic efficacy and toxicity of drugs that undergo metabolic N-acetylation (27, 28). Certain NAT1 polymorphisms also have been implicated as risk factors for a variety of human cancers (29-31). In addition to heritable influences on acetylation capacity, NAT activity levels can be affected by exogenous agents (32). N-Arylhydroxamic acid metabolites (Figure 1e) of aromatic amines, such as N-hydroxy-2-acetylaminofluorene (N-OH-2AAF), cause irreversible inactivation of NATs in vitro, and administration of N-OH-2-AAF to hamsters caused a substantial loss of hepatic NAT1 activity (33-36). Mass spectrometric analysis revealed that the mechanism of NAT inactivation in vitro by N-arylhydroxamic acids involves NAT-catalyzed deacetylation to produce an N-arylhydroxylamine, which undergoes oxidation to a nitrosoarene, followed by reaction of the nitroso compound with the catalytically essential and highly nucleophilic Cys68 of the enzyme to form a sulfinamide adduct (37, 38). Inactivation of intracellular NATs would be expected to compromise the important detoxification pathway of arylamine N-acetylation and potentially could contribute to an exacerbation of various untoward effects of environmental arylamines on human health. It is also likely that the putative function of NATs in the conjugation of N-arylhydroxylamines (Figure 1b) would be attenuated by nitrosoarene-mediated inactivation of the enzymes. In this paper, we report the results of an examination of the effect of the nitrosoarene metabolites of four toxicologically significant arylamines on human NAT1 activity in vitro and in human cells. We hypothesized that the nitrosoarene metabolites of primary arylamines that are acetylated efficiently by NAT1 would be potent inactivators of the enzyme, whereas the nitroso derivatives of arylamines that are less readily acetylated by NAT1 would be much less effective inactivators. We recently reported that the second order rate constants for the N-acetylation of 4-ABP and 2-aminoflourene by recombinant human NAT1 are 1270 and 4120 s-1 mM-1, respectively (39). In contrast, the corresponding rate constant for aniline is 113 s-1 mM-1, and for 2-methylaniline (o-toluidine), it is 5.2 s-1 mM-1. Thus, it was anticipated that 4-nitrosobiphenyl (4-NO-BP) and 2-nitrosofluorene (2-NO-F) would be potent, irreversible inactivators of NAT1 and that nitrosobenzene (NO-B) and 2-nitrosotoluene (2-NO-T) would be much less effective (Figure 2).
Caution: 4-NO-BP, 2-NO-F, NO-B, and 2-NO-T should be handled in accordance with NIH Guidelines for the Laboratory Use of Chemical Carcinogens (40). Materials and Methods. 4-NO-BP and 2-NO-F were synthesized as reported (37, 38). Recombinant human NAT1 was overexpressed and purified as described previously (41). Human NAT1 polyclonal antibody was provided by Prof. E. Sim (University of Oxford, Oxford, United Kingdom) (42). HeLa cells were obtained from American Type Culture Collection (Rockville, MD). Dulbecco’s modified Eagle’s medium (DMEM) was purchased from Mediatech (Herndon, VA). Fetal bovine serum(FBS)waspurchasedfromAtlantaBiologicals(Lawrenceville, GA). Phosphate-buffered saline (PBS), trypsin-ethylenediaminetetraacetic acid (EDTA), penicillin-streptomycin-fungizome, Lipofectamine 2000 transfection kit, 4-12% SDS-polyacrylamide gels, and PVDF membranes were obtained from Invitrogen (Carlsbad, CA). Goat antirabbit IgG horseradish peroxidase was obtained from Amersham Biosciences (Piscataway, NJ). Acetyl coenzyme A (AcCoA), antibeta actin, glyceraldehyde 3-phosphate, GSH, oxidized glutathione (GSSG), goat antirabbit IgG horseradish peroxidase, monoclonal antibeta EZview Red ANTIFLAG M2 Affinity Gel, NO-B, NADPH, p-aminobenzoic acid (PABA), p-nitrophenyl acetate (PNPA), pepsin A, and sulfamethazine (SMZ) were purchased from Sigma (St. Louis, MO). 2-NO-T was purchased from Aldrich (Milwaukee, WI). Restore Plus Western Blot Stripping Buffer was purchased from Pierce (Rockford, IL). Protein concentrations were determined with the Bradford assay (43). All buffers were degassed under vacuum, and all incubations were conducted under aerobic conditions. Spectrophotometric data were acquired on a Varian Cary 50 UV-vis spectrophotometer. Kinetic data were analyzed with the JMP IN software suite (SAS Institute, Inc.). IC50 and LC50 values were calculated with Kaleidagraph 3.6 (Synergy Software). NAT1 Activity Assay. The activity assay with human recombinant NAT1 was conducted as described previously (41). Time-Dependent Inactivation of NAT1 by Nitrosoarenes at 37 °C. The incubation mixtures contained NAT1 (35 µg/ mL, 1 µM), 4-NO-BP (0.1-5 µM), 2-NO-F (0.1-2 µM), NO-B (100-1250 µM), or 2-NO-T (100-1200 µM) and potassium phosphate buffer (20 mM, pH 7.4; 1 mM EDTA) in a total volume of 75 µL. The reaction was initiated by addition of nitrosoarene dissolved in DMSO (1.5 µL). The final concentration of DMSO was 2%. Aliquots (2 µL) were withdrawn at 15 (4-NO-BP, 2-NO-F), 30 (2-NO-T), or 60 s (NO-B) intervals and transferred to an assay cuvette. The controls contained DMSO but not nitrosoarene. Inactivation of NAT1 by NO-B and 2-NO-T in the Presence of AcCoA. In a total volume of 98 µL, NAT1 (35 µg/mL, 1 µM) was incubated with AcCoA (20 µM) in potassium phosphate buffer (20 mM, pH 7.4; 1 mM EDTA) at 37 °C. After 1 min, 2-NO-T or NO-B dissolved in DMSO (2 µL) was added. The final concentrations of 2-NO-T and NO-B were 300 and 250 µM, respectively. After incubation for 2 (2-NO-T) or 4 min (NO-B), aliquots (2 µL) were withdrawn and assayed for NAT1 activity. Inactivation of NAT1 by 4-NO-BP and 2-NO-F in the Presence of GSH. To NAT1 in potassium phosphate buffer (20 mM, pH 7.4; 1 mM EDTA) was added GSH dissolved in 1 µL of potassium phosphate buffer. After 2 min at 37 °C, either 4-NO-BP or 2-NO-F dissolved in DMSO (3 µL) was added. The final concentrations were as follows: NAT1, 25 µg/mL (0.72 µM); GSH, 0.5, 1, or 5 mM; 4-NO-BP or 2-NO-F, 1 µM; and
InactiVation of NAT1 by Nitrosoarenes
DMSO, 2%, in a total volume of 150 µL. After a 60 s incubation, aliquots (2 µL) were withdrawn and transferred to an assay cuvette. Controls contained only potassium phosphate buffer and DMSO. There was no significant difference between control activities determined in the presence and absence of GSH (p > 0.2). Sample Preparation for Nanoelectrospray Ionization (ESI)-Quadrupole-Time-of-Flight (Q-TOF) MS of 2-NOF-Treated NAT1. To NAT1 (120 µg, 43 µM) in potassium phosphate buffer (80 µL, 20 mM, pH 7.4; 1 mM EDTA; 10% glycerol; 0.1 mM DTT) was added 2-NO-F dissolved in 2 µL of DMSO. The final concentration of 2-NO-F was 250 µM. DMSO only was added to the control incubation mixtures. After a 5 min incubation at 37 °C, a portion (10 µL) of the reaction mixture was used to measure the residual activity. The reaction mixture was loaded onto a Bio Spin 6 Tris Column. After centrifugation, the sample was concentrated to 30 µL by freezedrying. Nano-ESI-Q-TOF MS of 2-NO-F-Treated NAT1. The NAT1 samples were desalted with Millipore C4 ZipTips according to the supplier’s protocol. ESI mass spectra were acquired as described (38). Pepsin Digestion of 2-NO-F-Treated NAT1. NAT1 was inactivated with 2-NO-F under conditions identical with those described above for the sample preparation for Nano-ESI-QTOF MS. The digestion was conducted as previously reported (38). MS Screening and Sequencing of Peptides. Peptides were desalted and analyzed by matrix-assisted laser desorption ionization (MALDI) Q-TOF MS and sequenced by MALDI Q-TOF MS/MS as previously reported (38). Cell Culture Conditions. HeLa cells were maintained as monolayers in DMEM supplemented with 10% FBS and 1% penicillin-streptomycin-fungizome in a humidified, 5% CO2 atmosphere at 37 °C. Western Blot Analysis of Endogenous NAT1 in HeLa Cells. At approximately 90% confluence, cell monolayers (100 mm plate, 3.45 × 106 cells) were washed with 5 mL of PBS. Cell monolayers were exposed to 2 mL of trypsin-EDTA and incubated for 3 min at 37 °C. Monolayers were washed with PBS buffer (10 mL) and centrifuged for 5 min at 1000g (4 °C). Cells were resuspended in 0.8 mL of lysis buffer [20 mM TrisHCl, pH 7.5; 1 mM EDTA; 0.1% Triton X-100; 1 mM phenylmethanesulfonyl fluoride (PMSF); 1 mM benzamidine; 1.4 µM pepstatin A; and 2.0 µΜ leupeptin] and disrupted at 4 °C by sonication (5 × 5 s bursts). The cell lysate was centrifuged for 15 min at 13000 rpm (4 °C), and the supernatant was withdrawn for analysis. The cell cytosol was subjected to electrophoresis on 4-12% SDS-polyacrylamide gels and transferred to PVDF membranes (30 V, 70 min, room temperature). Membranes were blocked with tris-buffered saline (TBS)/Tween 20 (0.05%) supplemented with 5% (w/v) nonfat milk powder. The membranes were incubated overnight at 4 °C with primary antibody (anti-NAT1, 1:4000) in TBS/Tween 20 supplemented with 5% (w/v) nonfat milk powder. The membranes were washed three times for 10 min with TBS/Tween 20. Secondary conjugated antibody (goat antirabbit IgG horseradish peroxidase, 1:4000) in TBS/Tween 20 supplemented with 5% (w/v) nonfat milk powder was added, and the membrane was incubated at room temperature for 1 h followed by four 10 min washes with TBS/Tween 20. The Enhanced Chemiluminescence Western Blotting analysis system (Amersham Biosciences, Piscataway, NJ) was used for detection. The membranes were scanned with a Chemifluorescence Workstation (Molecular Dynamics Storm
Chem. Res. Toxicol., Vol. 21, No. 10, 2008 2007
840, Sunnyvale, CA) and analyzed with Glyko BandScan Analysis Software (Glyko, Inc., Novato, CA), or the relative densities of the bands were analyzed with a Bio-Rad model GS700 Imaging Densitometer. The membranes were incubated with 20 mL of Restore Plus Western Blot Stripping Buffer for 15 min at room temperature to remove primary and secondary antibodies and washed with TBS/Tween 20 according the supplier’s protocol. Membranes were blocked with TBS/Tween 20 supplemented with 5% nonfat milk powder. The membranes were incubated with primary antibody (antibeta actin, 1:4000) in TBS/Tween 20 for 1 h and washed with TBS/Tween 20. Secondary conjugated antibody (goat antimouse IgG horseradish peroxidase, 1:4000) in TBS/ Tween 20 was added. After the membrane was incubated for 1 h and washed with TBS/Tween 20, the signal was detected and quantified as described above. NAT1 Activity in HeLa Cell Cytosol. Cell cytosol (50 µL) and PABA, dissolved in 40 µL of assay buffer (20 mM TrisHCl, 1 mM EDTA, pH 7.5), were incubated at 37 °C for 5 min. AcCoA dissolved in 10 µL of assay buffer was added. The final concentrations were as follows: PABA, 200 µM; AcCoA, 400 µM. The incubation was continued at 37 °C for various lengths of time (2-10 min). Cold trichloroacetic acid (20% w/v) (100 µL) was added, followed by centrifugation for 5 min at 12000g. The supernatant was added to 4-dimethylaminobenzaldehyde (800 µL, 5% w/v in 9:1 acetonitrile/water). The unreacted PABA was quantitated by measuring the absorbance at 450 nm (450nm ) 6400 M-1 cm-1) (44). All assays were performed in triplicate under initial rate conditions. The controls contained either assay buffer only, AcCoA without PABA, or PABA without AcCoA. When p-aminosalicylic acid (PAS) (200 µM) was used as acetyl acceptor, the unreacted PAS was quantitated by measuring the absorbance at 405 nm (405nm ) 1300 M-1 cm-1) (44). Glyceraldehyde 3-Phosphate Dehydrogenase (GAPDH) Activity in HeLa Cell Cytosol. Cell cytosol (20 µL), glyceraldehyde 3-phosphate (4 mM), and NAD+ (1 mM) were dissolved in 100 mM potassium phosphate buffer (10 mM EDTA, 0.1 mM DTT, pH 7.4). The final volume was 1 mL. NADH formation was monitored at 340 nm for 1min at 37 °C (45). Glutathione Reductase (GR) Activity in HeLa Cell Cytosol. Cell cytosol (40 µL) was incubated with 40 µL of 6 mM EDTA, 40 µL of 22 mM GSSG, and 240 µL of 180 mM sodium phosphate buffer (pH 7.2) for 5 min. The reaction was started by the addition of 40 µL of 4.5 mM NADPH. The rate of disappearance of NADPH was measured at 340 nm for 3 min at 37 °C (46). Effect of 4-NO-BP, 2-NO-F, NO-B, and 2-NO-T on NAT1 Activity in HeLa Cell Cytosol. In a total volume of 50 µL, cell cytosol (48 µL) was incubated with either 4-NO-BP (10-300 nM), 2-NO-F (10-300 nM), NO-B (50-750 µM), or 2-NO-T (25-1000 µM) at 37 °C for 10 min. The reaction was initiated by the addition of the nitrosoarenes dissolved in DMSO (2 µL). PABA and AcCoA dissolved in assay buffer (50 µL) and warmed to 37 °C for 5 min were added to the mixtures. The final concentrations were as follows: PABA, 200 µM; AcCoA, 400 µM. The residual NAT1 activity was measured as described above. The control incubations contained DMSO but not nitrosoarene. Effect of AcCoA on 4-NO-BP, 2-NO-F, NO-B, and 2-NO-T Inactivation of NAT1 in HeLa Cell Cytosol. To cell cytosol (46 µL) was added AcCoA dissolved in assay buffer (2 µL). The mixture was incubated for 5 min at 37 °C followed
2008
Chem. Res. Toxicol., Vol. 21, No. 10, 2008
by the addition of 4-NO-BP, 2-NO-F, NO-B, or 2-NO-T in DMSO (2 µL). The final concentrations were as follows: AcCoA, 400 µM; 4-NO-BP, 100 nM; 2-NO-F, 50 nM; NO-B, 500 µM; and 2-NO-T, 100 µM. After incubation for 10 min at 37 °C, PABA and AcCoA dissolved in assay buffer (50 µL) were added. The final concentrations were as follows: PABA, 200 µM; AcCoA, 400 µM. Residual NAT1 activity was measured as described above. For experiments without AcCoA, cell cytosol (46 µL) was incubated with 2 µL of assay buffer for 5 min. Nitrosoarenes (2 µL) were added, and incubations were continued for 10 min. PABA and AcCoA dissolved in 50 µL of assay buffer were added, and the NAT1 activity was measured. Effect of Nitrosoarenes on GAPDH Activity in HeLa Cell Cytosol. In a total volume of 50 µL, cell cytosol (48 µL) was incubated with 4-NO-BP (100-3000 nM), 2-NO-F (100-3000 nM), NO-B (200-4000 µM), or 2-NO-T (500-2000 µM) at 37 °C for 10 min. The reaction was initiated by the addition of the nitrosoarenes dissolved in DMSO (2 µL). A 20 µL portion of the incubation mixture was withdrawn to determine the residual GAPDH activity. The controls contained DMSO but not nitrosoarenes. Effect of Nitrosoarenes on GR Activity in HeLa Cell Cytosol. In a total volume of 100 µL, cell cytosol (96 µL) was incubated with 4-NO-BP (100-1500 nM), 2-NO-F (100-2000 nM), NO-B (800-5000 µM), or 2-NO-T (500-3000 µM) at 37 °C for 10 min. The reaction was initiated by the addition of the nitrosoarenes dissolved in DMSO (4 µL). A 40 µL portion of the incubation mixture was withdrawn for the GR activity assay. The controls contained DMSO but not nitrosoarenes. Determination of the Comparative Cytotoxicities of 4-NO-BP, 2-NO-F, NO-B, and 2-NO-T. Cell viability was determined by the 96 AQueous One Solution Cell Proliferation Assay (Promega Co., Madison, WI). HeLa cells were seeded into 96 well plates at a density of 6000 cells per well and allowed to adhere overnight. Media was changed with 100 µL of serum-free DMEM, which contained 4-NO-BP (final concentration 1-250 µM), 2-NO-F (final concentration 1-500 µM), NO-B (final concentration 1-500 µM), or 2-NO-T (final concentration 10-400 µM) dissolved in DMSO. DMSO only was added to the controls. The final concentration of DMSO was less than 1%. After a 4 h incubation at 37 °C, media was changed with 100 µL of serum-containing DMEM. After 40 h of growth, 20 µL of CellTiter 96 AQueous One Solution Reagent was added to each well. The plate was incubated at 37 °C for an additional 2 h. The absorbance at 490 nm was recorded with a FL600 Microplate Fluorescence Reader (Key Scientific, Mt. Prospect, IL). Effect of Nitrosoarenes on Endogenous NAT1, GAPDH, and GR Activities in HeLa Cells. At approximately 90% confluence, cell monolayers (100 mm plate, 3.45 × 106 cells) were washed twice with PBS. The cells were exposed to 4-NOBP (1-40 µM), 2-NO-F (0.5-20 µM), NO-B (100-250 µM), or 2-NO-T (50-200 µM) in 10 mL of serum-free DMEM media containing DMSO. The final concentration of DMSO was 0.1%. The incubation was continued at 37 °C for various periods of time. Cells were washed with PBS buffer, trypsinized, and harvested by centrifugation at 1000g for 5 min at 4 °C. The controls contained DMSO only. Cytosol was prepared as described above, and the NAT1, GAPDH, and GR activities were measured. After treatment with either 4-NO-BP or 2-NOF, cell viability was confirmed with the trypan blue assay. Cells were washed with PBS buffer and trypsinized, and 0.2 mL of the cell suspension was mixed with trypan blue solution (0.5
Liu et al.
mL, 0.4% w/v) (Sigma, St. Louis, MO) and 0.3 mL of Hanks’ balanced salts (HBSS). The mixture (10 µL) was used to count the viable cells with a standard hemocytometer according to the instructions from the supplier. Effect of N-Acetyl-L-Cysteine (NAC) on Inactivation of HeLa Cell NAT1 by 4-NO-BP. At approximately 90% confluence, cell monolayers were washed twice with PBS (5 mL). The cells were exposed to NAC (20 mM) in 10 mL of serumfree DMEM media for 4 h (47). The cell monolayers were washed five times with PBS (5 mL). Serum-free DMEM media (10 mL) containing 4-NO-BP (10 µM) dissolved in DMSO (10 µL) was added. The incubation was continued at 37 °C for 1 h. Cells were washed twice with PBS buffer (5 mL), trypsinized, and harvested by centrifugation at 1000g for 5 min (4 °C). Cytosol was prepared as described above, and the NAT1 activity was measured. Sample Preparation for In-Gel Digestion of FLAGNAT1 from 4-NO-BP-Treated p3FLAG-NAT1*4 Transfected HeLa Cells. At 90-95% confluence, cell monolayers (100 mm plate, 15 plates) were transfected with p3FLAGNAT1*4 plasmid with Lipofectamine 2000 (48). After 24 h, the media was replaced with serum-free media (10 mL) containing 4-NO-BP (200 µM) dissolved in DMSO (10 µL). After 1.5 h, the media was again replaced with serum-free media (10 mL) containing 4-NO-BP (200 µM) dissolved in DMSO (10 µL), and incubation was continued for 1.5 h. Cells were washed three times with PBS and were harvested. The controls contained DMSO but not 4-NO-BP. Cell cytosol (60 mg of protein from p3FLAG-NAT1*4 transfected HeLa cells; 59 mg of protein from 4-NO-BP treated p3FLAG-NAT1*4 transfected HeLa cells) was incubated with 300 µL (packed gel volume) of EZview Red ANTI-FLAG M2 Affinity Gel with shaking at 4 °C overnight. After centrifugation at 8200g for 30 s, the supernatant was removed. Gel pellets were washed three times with 5000 µL of TBS (50 mM Tris HCl, 150 mM NaCl, pH 7.4) containing 0.5 M NaCl for 10 min and twice with 5000 µL of TBS for 10 min. Nonreducing SDS-PAGE sample buffer (300 µL) was added to each sample. The samples were boiled for 10 min and centrifuged at 8200g for 3 min. The supernatant was loaded onto 4-12% SDSpolyacrylamide gels and subjected to electrophoresis. In-Gel Pepsin Digestion and MS Analysis of Overexpressed NAT1 in HeLa Cells after Treatment with 4-NOBP. The 3FLAG-NAT1 bands were cut from the gels and sliced into pieces. Gel pieces were washed with ddH2O (10 mL) for 60 min. Water was removed, and the gel pieces were destained with 5000 µL of 40 mM NH4HCO3/50% CH3CN at 37 °C for 30 min; the destaining was repeated twice. Gel pieces were dried with a vacuum centrifuge. The dry gel pieces were rehydrated with 800 µL of freshly prepared pepsin solution (50 µg/mL in H2O, pH 1.3). After incubation overnight at 37 °C, the pepsin solution was collected. Formic acid (5% v/v) in CH3CN:H2O (50% v/v) (500 µL) was added to the gel pieces, and the incubation was continued for 30 min; the procedure was carried out three times. All of the extracts were combined and dried in a centrivap concentrator. Samples were dissolved in 13 µL of reconstitution solution (CH3CN:H2O, 5:95, 0.5% TFA) and desalted with Millipore C18 ZipTips. Peptides were analyzed and sequenced as previously described (38).
Results Inactivation of Recombinant Human NAT1 by 4-NOBP, 2-NO-F, NO-B, and 2-NO-T. Incubation of human NAT1 with each of the nitrosoarenes caused a time-dependent loss of
InactiVation of NAT1 by Nitrosoarenes
Chem. Res. Toxicol., Vol. 21, No. 10, 2008 2009
Table 1. Kinetic Parameters for Inactivation of Human NAT1 by Nitrosoarenesa nitrosoarene 4-NO-BP NO-B 2-NO-F 2-NO-T
KI (µM)
kinact (s-1) kinact/KI (M-1 s-1) k2 (M-1 s-1)
0.67 ( 0.07 0.04 ( 0.001 540 ( 151 0.01 ( 0.002
59200 ( 7600 25 ( 12
34500 ( 1020 23 ( 2.0
a Results are expressed as the means ((SD) of three experiments. See the Supporting Information (Figure S1) for graphical illustrations of the kinetic analyses.
N-acetylation activity. The rate of NAT1 inactivation by both 4-NO-BP and NO-B was dependent on the concentration of nitrosoarene at lower concentrations, with saturation occurring at approximately 3 µM for 4-NO-BP and 1.0 mM for NO-B. In contrast, the rate of inactivation of NAT1 with both 2-NO-F and 2-NO-T increased linearly with increasing concentration of inactivator, and saturation was not observed within the range of concentrations that were studied. The plots of the logarithms of percentage residual activity vs incubation times for each concentration of each of the four nitrosoarenes were linear (Supporting Information, Figure S1), indicating that the inactivations were pseudo first-order processes; a first-order inactivation rate constant (kobs) for each concentration of nitrosoarene was obtained from the slopes of the lines generated by the plots. The kobs values obtained with 4-NO-BP and NO-B, each of which exhibited saturation kinetics, were analyzed according to eq 1, where [I] is the inhibitor concentration, to determine the dissociation constants (KI), first-order inactivation rate constants (kinact), and second-order inactivation rate constants (Table 1) (49).
kobs ) kinact ⁄ (1 + KI ⁄ [I])
(1)
The second-order inactivation rate constants (k2) for 2-NO-F and 2-NO-T, which did not exhibit saturation kinetics, were obtained by fitting the data to eq 2, where [I] is the inhibitor concentration and n is the order of the reaction with respect to inhibitor (50). For both 2-NO-F and 2-NO-T, the reaction order was 1.0, although the k2 value for 2-NO-F was 1500-fold greater than that of 2-NO-T (Table 1).
log kobs ) log k2 + n log [I]
(2)
As shown in Table 1, the second order rate constants for inactivation of NAT1 by 4-NO-BP and 2-NO-F are approximately 1500-2500-fold greater than the corresponding rate constants determined for NO-B and 2-NO-T. These results are consistent with the proposal that the nitrosoarene metabolites of arylamines that are efficiently acetylated by NAT1 can be expected to be more effective inactivators of the enzyme than the nitroso metabolites of arylamines that are less readily N-acetylated. MS Analysis of 2-NO-F-Treated NAT1. It was shown previously that the irreversible inactivation of human NAT1 by 4-NO-BP, as well as the inactivation of hamster NAT1 by 2-NOF, was the result of reaction of the nitroso substituent with the highly nucleophilic thiol of Cys68 to form a sulfinamide adduct (ArNHSO-NAT) (37, 38). To determine whether or not the inactivation of human NAT1 by 2-NO-F also involved sulfinamide adduct formation with the catalytically essential Cys68, NAT1 was incubated with 2-NO-F, and the adducted protein was analyzed by nano-ESI Q-TOF mass spectrometry. The mass spectrum of the adducted protein (Supporting Information, Figure S2) exhibited a minor peak (34300.3 Da), which corresponds to the native NAT1 mass of 34299.2 Da, as well
Figure 3. Effect of GSH on the inactivation of recombinant NAT1 (0.72 µM) by either 4-NO-BP (1 µM) or 2-NO-F (1 µM). The incubation time was 60 s. Each bar represents the mean and standard deviation of the results from three experiments. Asterisks represent significant differences from controls: *p < 0.05, **p < 0.01, and ***p < 0.001.
as a major peak (34494.9 Da), indicating the addition of a single 2-NO-F molecule (+195 Da). MALDI Q-TOF MS analysis of the pepsin digest of the 2-NO-F modified protein revealed a modified (+32 Da) peptide of mass 1590.80 Da, which was not present in the digest obtained with NAT1 that had not been treated with 2-NO-F (Supporting Information, Figure S3). It was found previously that protease digestion of NATs containing a sulfinamide adduct causes hydrolysis of the sulfinamide to a sulfinic acid (-SO2H), which accounts for a 32 Da increase in the mass of the modified peptide (37, 38). MALDI Q-TOF MS/ MS of the peptide of mass 1590.80 allowed identification of the modified (+32 Da) sequence DQVVRRNRGGWCL (Asp57Leu69; unmodified theoretical mass, 1558.78 Da). The intense b12-H2SO2 ion (1393.72 m/z), the b12 ion (1459.71 m/z), and the b11-NH3 ion (1307.68 m/z) verified the added mass of 32 Da and demonstrated that the side chain thiol of Cys68 had been converted to a sulfinic acid (Supporting Information, Figure S4). Effect of AcCoA on the Inhibition of NAT1 by NO-B and 2-NO-T. As shown in Table 1, NO-B and 2-NO-T were much less potent inactivators of recombinant NAT1 than either 4-NO-BP or 2-NO-F. Evidence that inactivation of the enzyme by NO-B and 2-NO-T involves interaction with the active site was obtained by incubation of the nitrosoarenes with NAT1 in the presence of the endogenous acetyl donor, AcCoA. Acetylation of the catalytically essential Cys68 would be expected to limit the ability of the nitrosoarenes to react with Cys68 and thereby protect the enzyme from inactivation. Incubation of NAT1 with NO-B (250 µM) caused 59 ((3)% inhibition of the enzyme, whereas the presence of AcCoA (20 µM) lowered the extent of inhibition to 13 ((4)%. Similarly, 2-NO-T (300 µM) inhibited NAT1 activity by 46 ((3)%, but in the presence of AcCoA (20 µM), the enzyme activity was inhibited only 5 ((5)%. Inactivation of NAT1 by 4-NO-BP and 2-NO-F in the Presence of GSH. The relatively facile reaction of GSH with various nitrosoarenes has been reported by a number of laboratories (21-23, 51-53). Thus, experiments were conducted to determine whether or not physiologically relevant concentrations of GSH would prevent the inactivation of NAT1 by either 4-NO-BP or 2-NO-F. In these experiments, the concentration of nitrosoarene in the incubation mixture was 1 µM, the ratio of nitrosoarene to NAT1 was 1.4:1, and the incubation time was 60 s. As shown in Figure 3, incubation with 4-NO-BP caused a 71% reduction in NAT1 activity and 2-NO-F caused
2010
Chem. Res. Toxicol., Vol. 21, No. 10, 2008
Liu et al.
Figure 4. Western blot analysis of recombinant NAT1 and HeLa cell NAT1 with a NAT1-specific antibody (42). Recombinant NAT1: lane 1, 0.25 ng; lane 2, 0.5 ng; lane 3, 1.0 ng; and lane 4, 1.5 ng. HeLa cell cytosolic protein: lane 6, 20 µg; lane 7, 30 µg; lane 8, 40 µg; and lane 9, 50 µg. The protein was quantified by densitometry.
Table 2. Inhibition of NAT1, GAPDH, and GR in HeLa Cell Cytosol IC50 (µM)a nitrosoarene 4-NO-BP 2-NO-F NO-B 2-NO-T a
NAT1
GAPDH
GR
0.06 ( 0.002 0.04 ( 0.003 237 ( 10 91 ( 16
1.0 ( 0.08 1.0 ( 0.09 1105 ( 78 402 ( 32
0.4 ( 0.03 0.8 ( 0.05 2033 ( 80 1418 ( 103
Results are expressed as the means ((SD) of three experiments.
91% inactivation. In the presence of a 500-fold excess of GSH (0.5 mM), treatment of NAT1 with 4-NO-BP resulted in a 35% loss of activity and 2-NO-F caused a 28% loss of activity. The presence of 1 mM GSH decreased the extent of inactivation by 4-NO-BP and 2-NO-F to 12 and 17%, respectively, and both nitrosoarenes caused about 10% loss of activity when incubated with NAT1 and 5 mM GSH. These results indicate that 4-NOBP and 2-NO-F react more readily with NAT1 than with GSH, even when GSH is present in much higher concentrations than either the enzyme or the nitrosoarene. NAT1 in HeLa Cells. NAT1 activity has been detected previously in HeLa cells (54).To verify the presence of NAT1 protein in HeLa cell cytosol, Western blot analysis was carried out with an antibody that specifically detects NAT1 (Figure 4) (42). Densitometric analysis and comparison with a standard curve obtained with recombinant NAT1 indicated the presence of 2.4 ((0.15) ng of NAT1 protein in 100 µg of cytosolic protein. N-Acetylation activity in HeLa cell cytosol was measured by using NAT1 substrates, PAS and PABA, with AcCoA as the acetyl donor. The specific activity with PAS was 4.08 ( 0.58 nmol/mg of protein/min, and with PABA, it was 4.4 ( 0.32 nmol/mg of protein/min. By comparison, the specific activity for PAS with homogeneous recombinant NAT1 was 337 ( 12 µmol/mg of protein/min, and the specific activity with PABA was 237 ( 2.4 µmol/mg of protein/min. On the basis of the specific activities obtained with PAS, HeLa cell cytosol contains 1.2 ng of NAT1 per 100 µg of protein, and on the basis of the specific activities obtained with PABA, the NAT1 concentration is 1.8 ng per 100 µg of protein. Thus, the cytosolic levels of NAT1 determined by either densitometric analysis or specific activities are similar and indicate that NAT1 constitutes approximately 0.002% of cytosolic protein in HeLa cells. It is of interest that the results of a partial purification indicated that NAT2 constitutes approximately 0.002% of soluble protein in human liver (55). Effect of 4-NO-BP, 2-NO-F, NO-B, and 2-NO-T on NAT1, GAPDH, and GR Activities in HeLa Cell Cytosol. HeLa cell cytosol was incubated with various concentrations of 4-NO-BP, 2-NO-F, NO-B, and 2-NO-T, and the IC50 values for inhibition of NAT1 were determined (Table 2). The IC50 values for inhibition of cytosolic GAPDH and GR by the four nitrosoarenes also were determined. GAPDH was selected for comparison with NAT1 because it is an abundant cytosolic enzyme that constitutes approximately 1% of HeLa cell protein
and because the catalytically essential cysteine thiolate of GAPDH has been found to react with a variety of electrophiles (56-58). GR was included in the study because it plays a critical role in maintenance of cellular redox homeostasis and because its active site cysteine thiols react with electrophilic reagents such as iodoacetamide and the decomposition products of nitrosoureas (59-61). Similar to the results obtained with recombinant NAT1 (Table 1), 4-NO-BP and 2-NO-F were 1600-5900 fold more potent than either NO-B or 2-NO-T as inhibitors of NAT1 in HeLa cell cytosol (Table 2). The IC50 values for inhibition of cytosolic GAPDH and GR by 4-NO-BP and 2-NO-F, however, ranged from 0.4 to 1.0 µM and were only 7-25-fold greater than the IC50 values for the inhibition of cytosolic NAT1 by the two nitrosoarenes. NO-B and 2-NO-T were much weaker inhibitors of cytosolic GAPDH and GR than either 4-NO-BP or 2-NO-F (Table 2). Effect of AcCoA on the Inhibition of HeLa Cell Cytosolic NAT1 Activity by Nitrosoarenes. Inclusion of AcCoA (400 µM) in the cytosol incubations with each of the four nitrosoarenes greatly diminished the extent of NAT1 inhibition. Incubation of cytosol with 4-NO-BP (100 nM) caused 64 ( 3% inhibition of NAT1 activity, but only 10 ( 8% inhibition occurred in the presence of AcCoA. 2-NO-F (50 nM) inhibited cytosolic NAT1 by 59 ( 5% and caused 10 ( 7% inhibition when AcCoA was present. Similarly, NO-B (500 µM) and 2-NO-T (100 µM) lowered the NAT1 activity by 62 ( 11 and 57 ( 7%, respectively, but only 7 ( 8 and 2 ( 5% inhibition was effected by NO-B and 2-NO-T when AcCoA was present. These results support the conclusion that inhibition of HeLa cell cytosolic NAT1 by all four nitrosoarenes involves interaction of the agents with the active site of the enzyme. Comparative Cytotoxicities of 4-NO-BP, 2-NO-F, NO-B, and 2-NO-T. To assess the relative cytotoxicities of the four nitrosoarenes, HeLa cells were plated at 6000 cells per well and were incubated with various concentrations of 4-NO-BP, 2-NO-F, NO-B, and 2-NO-T. Cell viability was determined with the MTS assay, and LC50 values were calculated (62). 4-NOBP and 2-NO-F, which were the most potent NAT1 inactivators (Tables 1 and 2), exhibited almost identical LC50 values of 40 ( 3 and 46 ( 3 µM, respectively. NO-B, with an LC50 of 273 ( 23 µM, was approximately 6-fold less cytotoxic to HeLa cells than either 4-NO-BP or 2-NO-F, and 2-NO-T was about 3.5-fold less toxic than either 4-NO-BP or 2-NO-F, having an LC50 value of 157 ( 5 µM. Effect of Nitrosoarenes on Intracellular NAT1, GAPDH, and GR Activities. To assess the effects of the nitrosoarenes on the intracellular activities of NAT1, GAPDH, and GR, HeLa cells were treated with a range of concentrations of the compounds for various lengths of time. Treatment of the cells with 4-NO-BP (2.5 µM) for 1 h caused a loss of 39% of the NAT1 activity, without causing a decrease in either GAPDH or GR activities (Figure 5A). Similarly, 4-NO-BP concentrations of 5 and 10 µM lowered the NAT1 activity by 42 and 53%, respectively, without affecting either GAPDH or GR. The
InactiVation of NAT1 by Nitrosoarenes
Figure 5. Concentration-dependent and time-dependent inactivation of HeLa cell NAT1, GAPDH, and GR by 4-NO-BP. (A) Concentration dependence. The incubation time was 1 h. Each bar represents the mean and standard deviation of the results of three experiments. Asterisks represent significant differences from control: *p < 0.05, **p < 0.02, and ***p < 0.001. (B) Time dependence. Upper panels: Western blot analysis of actin and NAT1 in 20 µg of cytosolic protein from HeLa cells that had been treated with 4-NO-BP (10 µM) for various periods of time. Protein was quantified by densitometry. Graph: The concentration of 4-NO-BP was 10 µM. Each bar represents the mean and standard deviation of the results of three experiments. Asterisks represent significant difference from control (0 min): *p < 0.001.
decreases in NAT1 activity caused by 4-NO-BP concentrations of 20 and 40 µM were almost identical at 60 and 59%, respectively. The 20 µM concentration of 4-NO-BP caused a reduction in GR activity to 82% of control values, but the 12% depression in GR activity that occurred in the presence of 40 µM 4-NOBP was not statistically significant (p < 0.05). At a concentration of 40 µM, 4-NO-BP lowered the GAPDH activity by only 14% (Figure 5A). These results demonstrate that intracellular NAT1 is highly susceptible to inhibition by 4-NO-BP, whereas the effect of low concentrations of 4-NO-BP on intracellular GAPDH or GR is negligible. The time dependence of inactivation of HeLa cell NAT1 was investigated by incubation of the cells with a 10 µM concentration of 4-NO-BP (Figure 5B). A 39% loss of NAT1 activity occurred within the first 15 min of exposure of the cells to 4-NOBP. The decreases in NAT1 activity at 30 and 60 min were 48 and 58%, respectively, with very little additional loss of activity observed after 2 h. It is of interest to note that more than half of the loss of NAT1 activity, measured after 2 h, had taken place within the first 15 min of exposure of the cells to 4-NOBP. In contrast, there was no inhibition of either GAPDH or GR during the 120 min incubation (Figure 5B). Western blot analysis was used to determine whether or not a decrease in NAT1 protein level occurred during treatment of HeLa cells with 4-NO-BP. As shown in Figure 5B, when HeLa
Chem. Res. Toxicol., Vol. 21, No. 10, 2008 2011
Figure 6. Concentration-dependent and time-dependent inactivation of HeLa cell NAT1, GAPDH, and GR by 2-NO-F. (A) Concentration dependence. The incubation time was 1 h. Each bar represents the mean and standard deviation of three experiments. Asterisks represent significant differences from controls: *p < 0.05, **p < 0.01, and ***p < 0.001. (B) Time dependence. The concentration of 2-NO-F was 10 µM. Each bar represents the mean and standard deviation of the results of three experiments. Asterisks represent significant differences from controls (0 min): *p < 0.05, **p < 0.01, and ***p < 0.001.
cell cytosol was probed with a human NAT1-specific antibody at various times during the course of exposure to 4-NO-BP (10 µM), there was no indication of a decrease in NAT1 protein, although a progressive loss of NAT1 activity occurred. The relative density of NAT1 protein bands was measured, and by use of a standard curve obtained with recombinant NAT1, it was determined that the average amount of NAT1 in the bands was 0.46 ( 0.04 ng. Butcher et al. also found that the loss of NAT1 activity caused by exposure of human peripheral blood mononuclear cells to N-hydroxy-PABA was not associated with a change in NAT1 protein levels (62). 2-NO-F was a somewhat more effective inactivator of intracellular NAT1 than 4-NO-BP. A 1 µM concentration of 4-NO-BP had no effect on NAT1 activity (Figure 5A), whereas treatment of the cells for 1 h with 1 µM 2-NO-F caused a 34% reduction in NAT1 activity (Figure 6A). Incubation with 2.5 and 5 µM concentrations of 2-NO-F caused 57% losses of NAT1 activity, and 10 µM 2-NO-F depressed activity by 70%, but no additional reduction was observed when the cells were exposed to a 20 µM concentration of 2-NO-F (Figure 6A). GAPDH and GR activities in HeLa cells were unaffected by 1 h treatments with 0.5-2.5 µM concentrations of 2-NO-F (Figure 6A). 2-NO-F concentrations of 5-20 µM caused minor decreases of 13-16% in GAPDH activity and 20-24% in GR activity. As shown in Figure 6B, 2-NO-F (10 µM) caused a 60% loss of cellular NAT1 activity in 15 min; after 30 min, the activity had been lowered by 73% and then remained constant through 120 min. After 120 min of incubation with 2-NO-F (10 µM), the GAPDH activity of the HeLa cells was 20% lower than the activity in untreated cells; the GR activity was 9% lower at 60
2012
Chem. Res. Toxicol., Vol. 21, No. 10, 2008
Liu et al.
Table 3. Effect of 4-NO-BP on NAT1 Activity after Exposure of HeLa Cells to NAC NAT1 (nmol/mg/min)a HeLa HeLa HeLa HeLa
cells cells + NAC cells + 4-NO-BP cells + NAC + 4-NO-BP
4.66 ( 0.11 4.54 ( 0.35 1.72 ( 0.25 1.61 ( 0.23
% control activity 100 97* 37** 35**
a Results are presented as the means ((SD) of three experiments. Asterisks represent significant differences from control values: *p > 0.05 and **p < 0.001.
min and 30% lower after 2 h. Thus, consistent with the results obtained with 4-NO-BP, the inhibitory effect of 2-NO-F on intracellular NAT1 activity is much more pronounced and has a much more rapid onset than the comparatively modest effects on GAPDH and GR activities. NO-B and 2-NO-T, which were relatively ineffective inhibitors of both recombinant NAT1 (Table 1) and cytosolic NAT1 (Table 2) were evaluated for their effects on HeLa cell NAT1, GAPDH, and GR activities. Incubation of cells for 6 h with NO-B (250 µM) did not cause a reduction in any of the three activities (data not shown). Similarly, treatment of HeLa cells for 4 h with 2-NO-T (100 µM) did not effect a change in NAT1, GAPDH, or GR activity (data not shown). Effect of N-Acetylcysteine (NAC) on the Inactivation of Intracellular NAT1 by 4-NO-BP. The GSH content of HeLa cells is reported to be 20-30 nmol per 106 cells (63, 64). Goswami et al. found that treatment of HeLa cells with NAC (20 mM) for 4 h resulted in the uptake of approximately 5 nmol of NAC per mg of cell protein and caused an increase in the ratio of reduced to oxidized GSH (47). Treatment of other human cell lines with NAC increased GSH levels by 3-4-fold and cysteine levels by 3-15-fold (65). To determine whether prior treatment of HeLa cells with NAC would influence the ability of 4-NO-BP to inactivate intracellular NAT1, cells were exposed to NAC (20 mM) for 4 h before being incubated with 4-NO-BP (10 µM) for 1 h. As shown in Table 3, 4-NO-BP caused a 63% reduction in the specific activity of HeLa cell NAT1, and the NAT1 activity was decreased by 65% when the cells had been treated with NAC before being exposed to the nitrosoarene. Thus, NAC had no effect on the inactivation of intracellular NAT1 by 4-NO-BP. Proteolysis and MALDI Q-TOF MS/MS Analysis of 4-NO-BP-Treated 3FLAG-NAT1 Expressed in HeLa Cells. To obtain evidence regarding the mechanism of inactivation of intracellular NAT1 by 4-NO-BP, 3FLAG-NAT1 was expressed in HeLa cells as previously reported (48). The specific activity of the NAT1 expressed in cell cytosol was 472 ( 32 nmol/mg of protein/min, which was more than 100-fold greater than the specific activity of endogenous NAT1. Incubation of the cells with 4-NO-BP (200 µM) for 3 h caused a reduction in NAT1 activity to 19 ( 6% of control values; cell viability was confirmed with the trypan blue assay. Immunoprecipitation of 3FLAG-NAT1 with anti-FLAG M-2 affinity gel, followed by release of the bound protein and in-gel proteolysis with pepsin, afforded digests that were analyzed by MALDI Q-TOF MS. The mass spectrum of peptides obtained from cells that had not been treated with 4-NO-BP contained a peak of mass 1558.79 Da, corresponding to the sequence DQVVRRNRGGWCL (Asp57-Leu69; unmodified theoretical mass 1558.78 Da) (Supporting Information, Figure 5). In contrast, after treatment of cells with 4-NO-BP, the corresponding peptide mass spectrum contained a new peak at 1590.76 Da, which corresponds to the modified (+32 Da) Asp57-Leu69 sequence (Supporting Infor-
mation, Figure 5). MALDI Q-TOF MS/MS of the peptide of mass 1590.76 revealed both a strong b12-H2SO2 ion (1393.71 m/z) and a b12 ion of 1459.71 (m/z), which confirms that the thiol group of Cys68 is present in the sulfinic acid form. All other b and y ions in the spectrum were consistent with the Cys68 side chain having been converted to a sulfinic acid (Figure 7). These results support the conclusion that the intracellular inactivation of NAT1 by 4-NO-BP involves formation of a sulfinamide adduct with the catalytically essential Cys68 (Figure 8) (37, 38).
Discussion The interrelationships between variable NAT activities and the toxicity or carcinogenicity of environmental arylamines, as well as the influence of NAT activity levels on the efficacy and toxicity of therapeutic agents that contain arylamine and hydrazine functionalities, have been subject to extensive and ongoing investigation (27, 28, 66, 67). More recently, the potential for direct modulation of NAT activities by exogenous or endogenous chemical agents has attracted attention (32). Physiologically relevant concentrations of hydrogen peroxide cause a reversible inactivation of human NAT1 in vitro and also reversibly inactivate both NAT1 and NAT2 activities in lens epithelial cells (68, 69). Another endogenous oxidant, peroxynitrite, catalyzes the irreversible inactivation of NAT1 in MCF7 breast cancer cells and causes inactivation of both NAT1 and NAT2 activities in human lens epithelial cells and in mouse skeletal muscle cells (69-71). Of particular relevance to the study described in this paper are the findings of Butcher et al., who reported that incubation of the hydroxylamine of PABA (N-hydroxy-PABA) with peripheral blood mononuclear cells caused inactivation of NAT1; it was suggested that the nitroso oxidation product, formed from the hydroxylamine, could be responsible for inactivation of the enzyme (62). This suggestion is consistent with the observation of King and Duffel, who reported that inactivation of rat aryl sulfotransferase IV by N-hydroxyarylamines was dependent upon oxidation of the hydroxylamines to nitrosoarenes (72). Previously, we reported that treatment of hamster NAT1, hamster NAT2, and human NAT1 with 4-NOBP resulted in irreversible inactivation of the enzyme through formation of a sulfinamide adduct with the thiol group of the catalytic Cys68 residue and that a similar sulfinamide adduct is formed upon exposure of hamster NAT1 to 2-NO-F (37, 38). In this report, we confirmed by mass spectrometric analysis that inactivation of human NAT1 by 2-NO-F also is mediated by production of a covalent sulfinamide adduct with Cys68. A kinetic analysis of the inactivation of human NAT1 by 4-NO-BP, 2-NO-F, NO-B, and 2-NO-T was undertaken to test the hypothesis that nitrosoarene derivatives of primary arylamines that are rapidly N-acetylated by NAT1 should be potent inactivators of the enzyme. It was anticipated that both 4-NOBP and 2-NO-F would be potent, irreversible inhibitors and that NO-B and 2-NO-T, which are oxidation products of much less effective NAT1 substrates, would be less effective inhibitors. The inactivation of NAT1 in vitro by both 4-NO-BP and 2-NO-F was extremely rapid. 4-NO-BP (5 µM) reduced NAT1 activity to approximately 10% of control values within 60 s, and 2-NO-F (2 µM) caused a 95% loss of activity within 45 s. NO-B and 2-NO-T caused similar extents of inactivation of NAT1 as 4-NO-BP and 2-NO-F, but much higher concentrations and longer incubation times were required for NO-B and 2-NO-T (Supporting Information, Figure S1). The second order rate constants for inactivation of NAT1 by 4-NO-BP and 2-NO-F
InactiVation of NAT1 by Nitrosoarenes
Chem. Res. Toxicol., Vol. 21, No. 10, 2008 2013
Figure 7. MALDI Q-TOF tandem mass spectrum of the 1590.76 Da peptide obtained by pepsin digestion of NAT1 after overexpression of NAT1 in HeLa cells and treatment of the cells with 4-NO-BP. The asterisk indicates Cys68, which contains a sulfinic acid side chain.
Figure 8. Reaction of a nitrosoarene with the catalytically essential Cys68 of NAT1 to form a sulfinamide.
were 59200 and 34500 M-1 s-1, respectively, and exceeded those determined for NO-B and 2-NO-T by 1500-2500-fold (Table 1). The reported rate constant for inactivation of NAT1 by hydrogen peroxide is 7 M-1 s-1, and for the anticancer agent, cisplatin, it is 12 M-1 s-1 (68, 73). The inactivation rate constants for hydrogen peroxide and cisplatin are 2900-8000fold smaller than those of 4-NO-BP and 2-NO-F and are similar in magnitude to the values determined for NO-B and 2-NO-T. In contrast, the inactivation rate constant determined for the highly reactive oxidant, peroxynitrite, was 50000 M-1 s-1 and is similar to those of 4-NO-BP and 2-NO-F (70). GSH is an essential cellular reductant and a nucleophile that reacts with numerous nitrosoarenes to form sulfinamides (21-23, 51-53). To determine whether GSH could prevent the inactivation of NAT1 by 4-NO-BP and 2-NO-F, experiments were designed in which approximately equal concentrations of NAT1 and a nitrosoarene were incubated with a 500-5000fold excess of GSH for 60 s. In the presence of a 500-fold excess of GSH, 4-NO-BP and 2-NO-F caused substantial losses of NAT1 activity, and neither a 1000-fold nor a 5000-fold excess of GSH provided complete protection of the enzyme. Although rate constants have not been reported for the reaction of GSH with either 4-NO-BP or 2-NO-F, the rate constant for reaction of GSH with the chemically similar nitrosoarene, NO-B, is 5000 M-1 s-1 (23, 52). On the basis of the magnitude of their NAT1 inactivation rate constants and the extent of NAT1 inactivation that occurred in the presence of GSH, it can be concluded that both 4-NO-BP and 2-NO-F react much more readily with NAT1 than with GSH. NO-B and, presumably, 2-NO-T react with GSH more rapidly than with NAT1. Western blot experiments and specific activity comparisons with purified recombinant enzyme confirmed that NAT1 is a low abundance protein in HeLa cells. NAT1 constituted 0.002% of the soluble protein in HeLa cells, which is nearly identical to the calculated NAT2 level in human liver (55). In related experiments, it was found that two breast cancer cell lines,
MDA-MB-231 and MDA-MB-468, had NAT1 protein and specific activity levels that were almost identical with those of HeLa cells, whereas in the estrogen receptor-positive MCF7 breast cancer cell line, specific activity and densitometry measurements revealed that NAT1 constituted 0.005-0.01% of cytosolic protein (data not shown). The inhibitory effects of the nitrosoarenes on NAT1 activity in intact HeLa cells and in cell cytosol were consistent with the results obtained with recombinant NAT1 in that low concentrations of both 4-NO-BP and 2-NO-F effected a rapid loss of NAT1 activity, whereas NO-B and 2-NO-T were relatively weak inhibitors of cytosolic NAT1 and had no effect on cellular NAT1. NO-B and 2-NO-T also were ineffective inhibitors of cytosolic GADPH and GR. 4-NO-BP and 2-NOF, however, exhibited IC50 values of 0.4-1.0 µM as inhibitors of cell-free GAPDH and GR but had much less effect on the endogenous HeLa cell enzymes. Treatment of the cells with low concentrations of 4-NO-BP caused negligible inhibition of endogenous GAPDH and GR, and a low level of inhibition was observed with 2-NO-F only after 1-2 h of exposure or after treatment of the cells with the highest concentrations tested. It is likely that the intracellular concentrations achieved after exposure of the cells to lower concentrations of 4-NO-BP and 2-NO-F were insufficient to inhibit GAPDH and GR, although a marked reduction in NAT1 activity occurred. Approximately 30-40% of intracellular NAT1 was not susceptible to inactivation by either 4-NO-BP or 2-NO-F. Concentrations of 20 and 40 µM 4-NO-BP produced identical reductions of 60% in cellular NAT1 activity, and both 10 and 20 µM concentrations of 2-NO-F caused a 70% loss of activity. With both 4-NO-BP and 2-NO-F, a maximal loss of activity occurred within the first hour of exposure, and no additional reduction was measurable after 2-4 h. N-Hydroxy-PABA, in concentrations of 50-100 µM, also caused extensive but incomplete inactivation of NAT1 in peripheral blood mononuclear cells after exposures of 4-24 h, and treatment of two
2014
Chem. Res. Toxicol., Vol. 21, No. 10, 2008
types of human breast cancer cells for 1 h with 250 and 500 µM concentrations of cisplatin resulted in similar reductions in NAT1 activity of approximately 60-80% (62, 73). Because nitrosoarenes inactivate NAT1 through irreversible adduction of the active site Cys68, the residual NAT1 activity may represent a portion of the intracellular enzyme in which Cys68 is not available for reaction because it is in the acetylated state or, possibly, because the active sites of a portion of intracellular NAT1 are not accessible to the nitrosoarenes. It was reported that a 10 min exposure of MCF7 breast cancer cells to peroxynitrite (250 µM) caused almost complete loss of NAT1 activity, but because of the ability of peroxynitrite to oxidize and nitrate amino acid side chains, it is possible that at least part of the extensive inactivation of intracellular NAT1 by peroxynitrite is due to reaction with amino acid residues other than Cys68 (70, 74). Both 4-NO-BP and 2-NO-F were more effective inactivators of HeLa cell NAT1 and were more cytotoxic than either NO-B or 2-NO-T, but it is not known whether cytotoxicity is directly influenced by the reduction in NAT1 activity. Although the activities of HeLa cell GAPDH and GR were not affected by concentrations of 4-NO-BP and 2-NO-F that caused substantial losses of NAT1 activity, it is possible, if not likely, that the function of other cellular proteins may be altered either directly or indirectly by the nitosoarenes. Indeed, it has been suggested that arylamine cytotoxicity in rat hepatocytes is ultimately due to the effects of nitrosoarenes (75). Low concentrations of 2-NO-F uncouple oxidative phosphorylation in isolated mitochondrial preparations, and higher concentrations react with double bonds in microsomal membrane lipids to produce nitroxyl free radicals (76, 77). The nitroso metabolites of the arylamine antibacterial agents, sulfamethoxazole and dapsone, cause both protein haptenation and cytotoxicity (78, 79). Regardless of the mechanism(s) whereby various nitrosoarenes cause cytotoxicity, the results presented herein demonstrate that NAT1 is a preferred target of 4-NO-BP and 2-NO-F and that intracellular NAT1 is inactivated by concentrations of 4-NOBP and 2-NO-F that are not toxic to HeLa cells. The rapid, active site-directed inactivation of such a low abundance enzyme can be attributed to favorable interaction of the biphenyl and fluorenyl ring systems with hydrophobic residues in the NAT1 binding cavity, as well as to the enhanced nucleophility of the Cys68 side chain, which functions as the unprotonated thiolate in an imidazolium-thiolate ion pair (80-82). Human exposure to nitrosoarenes results not only from oxidation of environmental and therapeutic arylamines but as well from metabolic reduction of nitroarenes, such as 2-nitrofluorene and 1-nitropyrene, which are environmental byproducts of combustion (83, 84). Drugs that contain aromatic nitro groups also are a source of exposure to nitrosoarenes (85). Because of the multiplicity of potential sources of exposure to their chemical precursors, nitrosoarenes may have a substantial impact on the propensity to either detoxify or bioactivate arylamines through NAT1-mediated processes and to influence other physiological or pathological functions of the enzyme (86, 87). Acknowledgment. We are grateful to Professor Edith Sim, Oxford University, for the generous gift of the NAT1 antibody and to Fen Liu, University of Minnesota, and Professor Douglas Spitz, University of Iowa, for helpful discussions and advice. The mass spectrometry data were obtained with the assistance of Drs. Lorraine Anderson, Lee Ann Higgins, and Sean Murray of the Center for Mass Spectrometry and Proteomics, University of Minnesota. This research was supported in part by a
Liu et al.
Developmental Grant for Drug Design and Discovery from the Department of Medicinal Chemistry, University of Minnesota. Supporting Information Available: Plots of time- and concentration-dependent inactivation of recombinant human NAT1 by 4-NO-BP, 2-NO-F, NO-B, and 2-NO-T (Figure S1), nano-ESI-Q-TOF mass spectrum of recombinant human NAT1 after incubation with 2-NO-F (Figure S2), segments of the MALDI Q-TOF mass spectra of pepsin digests of native and 2-NO-F-inactivated recombinant human NAT1 (Figure S3), MALDI Q-TOF tandem mass spectrum of the 1590.80 Da peptide obtained by pepsin digestion of 2-NO-F-inactivated recombinant human NAT1 (Figure S4), and segments of the MALDI Q-TOF mass spectra of in-gel pepsin digests of NAT1 after overexpression in HeLa cells and of NAT1 after overexpression in HeLa cells followed by treatment of the cells with 4-NO-BP (Figure S5). This material is available free of charge via the Internet at http://pubs.acs.org.
References (1) Palmiotto, G., Pieraccini, G., Moneti, G., and Dolara, P. (2001) Determination of the levels of aromatic amines in indoor and outdoor air in Italy. Chemosphere 43, 355–361. (2) Richter, E., Rosler, S., Scherer, G., Gostomzyk, J. G., Grubl, A., Kramer, U., and Behrendt, H. (2001) Haemoglobin adducts from aromatic amines in children in relation to area of residence and exposure to environmental tobacco smoke. Int. Arch. Occup. EnViron. Health 74, 421–428. (3) DeBruin, L. S., Pawliszyn, J. B.and., and Josephy, P. D. (1999) Detection of monocyclic aromatic amines, possible mammary carcinogens, in human milk. Chem. Res. Toxicol. 12, 78–82. (4) Parkes, H. G., and Evans, A. E. J. (1984) Epidemiology of aromatic amine cancers. In Chemical Carcinogens, 2nd ed. (Searle, C. E., Ed.) Vol. 1, pp 277-301, American Chemical Society, Washington, DC. (5) Garner, R. C., Martin, C. N., and Clayson, D. B. (1984) Carcinogenic aromatic amines and related compounds. In Chemcal Carcinogens, 2nd ed. (Searle, C. E., Ed.) Vol. 1, pp 175-276, American Chemical Society, Washington, DC. (6) Miller, J. A. (1998) The metabolism of xenobiotics to reactive electrophiles in chemical carcinogenesis and mutagenesis: A collaboration with Elizabeth Cavert Miller and our associates. Drug. Metab. ReV. 30, 645–674. (7) Hanna, P. E. (1996) Metabolic activation and detoxification of arylamines. Curr. Med. Chem. 3, 195–210. (8) Cohen, S. M., Boobis, A. R., Meek, M. E., Preston, R. J., and McGregor, D. B. (2006) 4-Aminobiphenyl and DNA reactivity: Case study within the context of the 2006 IPCS Human Relevance Framework for Analysis of a cancer mode of action for humans. Crit. ReV. Toxicol. 36, 803–819. (9) Skipper, P. L., Tannenbaum, S. R., Ross, R. K., and Yu, M. C. (2003) Nonsmoking-related arylamine exposure and bladder cancer risk. Cancer. Epidemiol. Biomarkers PreV. 12, 503–507. (10) Gan, J., Skipper, P. L., Gago.Dominquez, M., Arakawa, K., Ross, R. K., Yu, M. C., and Tannenbaum, S. R. (2004) Alkylanilinehemoglobin adducts and risk of non-smoking related bladder cancer. J. Natl. Cancer Inst. 96, 1425–1431. (11) Airoldi, L., Orsi, F., Magagnotti, C., Coda, R., Randone, D., Casetta, G., Peluso, M., Hautefeuille, A., Malaveille, C., and Vineis, P. (2002) Determinants of 4-aminobiphenyl-DNA adducts in bladder cancer biopsies. Carcinogenesis 23, 861–866. (12) Zayas, B., Stillwell, S. W., Wishnok, J. S., Trudel, L. J., Skipper, P., Yu, M. C., Tannenbaum, S. R., and Wogan, G. N. (2007) Detection and quantification of 4-ABP adducts in DNA from bladder cancer patients. Carcinogenesis 28, 342–349. (13) Patrianakos, C., and Hoffman, D. (1979) Chemical studies on tobacco smoke LXIV. On the analysis of aromatic amines in tobacco smoke. J. Anal. Toxicol. 3, 150–154. (14) Smith, C. J., Dooly, G. L., and Moldoveanu, S. C. (2003) New technique using solid-phase extraction for the analysis of aromatic amines in mainstream cigarette smoke. J. Chromatogr. A 991, 99– 107. (15) Mommsen, S., and Aagaard, J. (1983) Tobacco as a risk factor in bladder cancer. Carcinogenesis 4, 335–338. (16) Bryant, M. S., Skipper, P. L., Tannenbaum, S. R., and Maclure, M. (1987) Hemoglobin adducts of 4-aminobiphenyl in smokers and nonsmokers. Cancer Res. 47, 602–608.
InactiVation of NAT1 by Nitrosoarenes (17) Sarkar, M., Stabbert, R., Kinser, R. D., Oey, J., Rustemeier, K., von.Holt, K., Schepers, G., Walk, R. A., and Roethig, H. J. (2006) CYP1A2 and NAT2 phenotyping and 3-aminobiphenyl and 4-aminobiphenyl hemoglobin adduct levels in smokers and non-smokers. Toxicol. Appl. Pharmacol. 213, 198–206. (18) Kim, D., and Guengerich, F. P. (2005) Cytochrome P450 activation of arylamines and heterocyclic amines. Annu. ReV. Pharmacol. Toxicol. 45, 27–49. (19) Beland, F. A., and Kadlubar, F. F. (1990) Metabolic activation and DNA adducts of aromatic amines and nitroaromatic hydrocarbons. Handbook Exp. Pharmacol. 94/I, 267–325. (20) Grantham, P. H., Weisburger, E. K., and Weisburger, J. H. (1965) Dehydroxylation and deacetylation of N-hydroxy-N-2-fluorenylacetamide by rat liver and brain homogenates. Biochim. Biophys. Acta 107, 414–424. (21) Mulder, G. J., Unruh, L. E., Evans, F. E., Ketterer, B., and Kadlubar, F. F. (1982) Formation and identification of glutathione conjugates from 2-nitrosofluorene and N-hydroxy-2-aminofluorene. Chem.-Biol. Interact. 39, 111–127. (22) Eyer, P. (1994) Reactions of oxidatively activated arylamines with thiols: reaction mechanisms and biologic implications. An overview. EnViron. Health Perspect. 102 (S6), 123–132. (23) Kazanis, S., and McClelland, R. A. (1992) Electrophilic intermediate in the reaction of glutathione and nitrosoarenes. J. Am. Chem. Soc. 114, 3052–3059. (24) Skipper, P. L., and Stillwell, W. G. (1994) Aromatic amine-hemoglobin adducts. Methods Enzymol. 231, 643–649. (25) Josephy, P. D., and Mannervik, B. (2006) Molecular Toxicology, 2nd ed., pp 426-447, Oxford University Press, New York. (26) Hanna, P. E. (1994) N-Acetyltransferases, O-acetyltransferases and N,O-acetyltransferases: Enzymology and bioactivation. AdV. Pharmacol. 27, 401–430. (27) Hein, D. W. (2006) N-Acetyltransferase 2 genetic polymorphism: effects of carcinogen and haplotype on urinary bladder cancer risk. Oncogene 25, 1649–1658. (28) Meisel, P. (2002) Arylamine N-acetyltransferases and drug response. Pharmacogenomics 3, 349–366. (29) Minchin, R. F., Hanna, P. E., Dupret, J. M., Wagner, C. R., RodriguesLima, F.and., and Butcher, N. J. (2007) Arylamine N-acetyltransferase I. Int. J. Biochem. Cell Biol. 39, 1999–2005. (30) Sim, E., Westwood, I., and Fullam, E. (2007) Arylamine N-acetyltransferases. Expert Opin. Drug Metab. Toxicol. 3, 169–184. (31) Walraven, J. M., Trent, J. O., and Hein, D. W. (2008) Structurefunction analyses of single nucleotide polymorphisms in human N-acetyltransferase 1. Drug Metab. ReV. 40, 169–184. (32) Dupret, J. M., and Rodrigues-Lima, F. (2005) Structure and regulation of the drug-metabolizing enzymes arylamine N-acetyltransferases. Curr. Med. Chem. 12, 311–318. (33) Hanna, P. E., Banks, R. B., and Marhevka, V. C. (1982) Suicide inactivation of hamster hepatic arylhydroxamic acid N,O-acyltransferase. A selective probe of N-acetyltransferase multiplicity. Mol. Pharmacol. 21, 159–165. (34) Wick, M. J., and Hanna, P. E. (1990) Bioactivation of N-arylhydroxamic acids by rat hepatic N-acetyltransferase. Detection of multiple enzyme forms by mechanism based inactivation. Biochem. Pharmacol. 39, 991–1003. (35) Sticha, K. R. K., Bergstrom, C. P., Wagner, C. R., and Hanna, P. E. (1998) Characterization of hamster recombinant monomorphic and polymorphic arylamine N-acetyltransferases. Bioactivation and mechanism based inactivation studies with N-hydroxy-2-acetylaminofluorene. Biochem. Pharmacol. 56, 47–59. (36) Smith, T. J., and Hanna, P. E. (1988) Hepatic N-acetyltransferases: Selective inactivation in vivo by a carcinogenic N-arylhydroxamic acid. Biochem. Pharmacol. 37, 427–434. (37) Guo, Z., Wagner, C. R., and Hanna, P. E. (2004) Mass spectrometric investigation of the mechanism of inactivation of hamster arylamine N-acetyltransferase 1 by N-hydroxy-2-acetylaminofluorene. Chem. Res. Toxicol. 17, 275–286. (38) Wang, H., Wagner, C. R., and Hanna, P. E. (2005) Irreversible inactivation of arylamine N-acetyltransferases in the presence of N-hydroxy-4-acetylaminobiphenyl: A comparison of human and hamster enzymes. Chem. Res. Toxicol. 18, 183–197. (39) Liu, L., Von Vett, A., Zhang, N., Walters, K. J., Wagner, C. R., and Hanna, P. E. (2007) Arylamine N-acetyltranserases: Characterization of the substrate specificities and molecular interactions of environmental arylamines with human NAT1 and NAT2. Chem. Res. Toxicol. 20, 1300–1308. (40) NIH (1981) NIH Guidelines for the Laboratory Use of Chemical Carcinogens, NIH Publication No. 81-2385, U.S. Government Printing Office, Washington, DC. (41) Wang, H., Vath, G. M., Kawamura, A., Bates, C. A., Sim, E., Hanna, P. E., and Wagner, C. R. (2005) Overexpression, purification, and
Chem. Res. Toxicol., Vol. 21, No. 10, 2008 2015
(42)
(43) (44)
(45) (46)
(47)
(48)
(49) (50) (51)
(52) (53) (54)
(55) (56)
(57)
(58) (59) (60) (61)
(62) (63)
(64)
characterization of recombinant human arylamine N-acetyltransferase 1. Protein J. 24, 65–77. Stanley, L. A., Coroneos, E., Cuff, R., Hickman, D., Ward, A., and Sim, E. (1996) Immunochemical detection of arylamine N-acetyltransferase in normal and neoplastic bladder. J. Histochem. Cytochem. 44, 1059–1067. Bradford, M. M. (1976) A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal. Biochem. 72, 248–254. Sinclair, J. C., Delgoda, R., Noble, M. E., Jarmin, S., Goh, N. K., and, and Sim, E. (1998) Purification, characterization, and crystallization of an N-hydroxyarylamine O-acetyltransferase from Salmonella typhimurium. Protein Expression Purif. 12, 371–380. McAlister, L., and Holland, M. J. (1985) Differential expression of the three yeast glyceraldehyde-3-phosphate dehydrogenase genes. J. Biol. Chem. 260, 15019–15027. Cohen, M. B., and Duvel, D. L. (1988) Characterization of the inhibition of glutathione reductase and the recovery of enzyme activity in exponentially growing murine leukemia (L1210) cells treated with 1,3-bis(2-chloroethyl)-1-nitrosourea. Biochem. Pharmacol. 37, 3317– 3320. Goswami, P. C., Sheren, J., Albee, L. D., Parsian, A., Sim, J. E., Ridnour, L. A., Higashikubo, R., Gius, D., Hunt, D. R., and Spitz, D. R. (2000) Cell cycle-coupled variation in topoisomerase IIR mRNA is regulated by the 3′-untranslated region. J. Biol. Chem. 275, 38384– 38392. Liu, F., Zhang, N., Zhou, X., Hanna, P. E., Wagner, C. R., Koepp, D. M., and Walters, K. J. (2006) Arylamine N-acetyltransferase aggregation and constitutive ubiquitylation. J. Mol. Biol. 361, 482– 492. Kitz, R., and Wilson, I. B. (1962) Esters of methanesulfonic acid as irreversible inhibitors of acetylcholinesterase. J. Biol. Chem. 237, 3245–3249. Levy, H. M., Leber, P. D., and Ryan, E. M. (1963) Inactivation of myosin by 2,4-dinitrophenol and protection by adenosine triphosphate and other phosphate compounds. J. Biol. Chem. 238, 3654–3659. Lotlikar, P. D., Miller, E. C., Miller, J. A., and Margreth, A. (1965) The enzymatic reduction of the N-hydroxy derivatives of 2-acetylaminofluorene and related carcinogens by tissue preparations. Cancer Res. 25, 1743–1751. Eyer, P. (1979) Reactions of nitrosobenzene with reduced glutathione. Chem.-Biol. Interact. 24, 227–239. Dolle, B., Topner, W., and Neumann, H.-G. (1980) Reaction of arylnitroso compounds with mercaptans. Xenobiotica 10, 527–536. Butcher, N. J., Tetlow, N. L., Cheung, C., Broadhurst, G. M., and Minchin, R. F. (2007) Induction of human arylamine N-acetyltransferase type I by androgens in human prostate cancer cells. Cancer Res. 67, 85–92. Grant, D. M., Lottspeich, F., and Meyer, U. A. (1989) Evidence for two closely related isozymes of arylamine N-acetyltransferase in human liver. FEBS Lett. 244, 203–207. Nakano, M., Funayama, S., de Oliveira, M. B. M., Sung, L. B., and Gomes, E. M. (1992) D-Glyceraldehyde-3-phosphate dehydrogenase from HeLa cells. 1. Purification and properties of the enzyme. Comp. Biochem. Physiol., Part B: Biochem. Mol. Biol. 102, 873–877. Dietze, E. C., Schafer, A., Omichinski, J. G., and Nelson, S. D. (1997) Inactivation of glyceraldehyde-3-phosphate dehydrogenase by a reactive metabolite of acetaminophen and mass spectral characterization of an arylated active site peptide. Chem. Res. Toxicol. 10, 1097–1103. Dennehy, M. K., Richards, K. A. M., Wernke, G. R., Shyr, Y., and Liebler, D. C. (2006) Cytosolic and nuclear protein targets of thiolreactive electrophiles. Chem. Res. Toxicol. 19, 20–29. Pai, E. F., and Schulz, G. E. (1983) The catalytic mechanism of glutathione reductase as derived from x-ray diffraction analyses of reaction intermediates. J. Biol. Chem. 258, 1752–1757. Babson, J. R., and Reed, D. J. (1978) Inactivation of glutathione reductase by 2-chloroethyl nitrosourea-derived isocyanates. Biochem. Biophys. Res. Commun. 83, 754–762. Karplus, P. A., Krauth-Siegel, R. L., Schirmer, R. H., and Schulz, G. E. (1988) Inhibition of human glutathione reductase by the nitrosourea drugs 1,3-bis(2-chloroethyl)-1-nitrosourea and 1-(2-chloroethyl)-3-(2-hydroxyethyl)-1-nitrosourea. Eur. J. Biochem. 171, 193– 198. Butcher, N. J., Ilett, K. F., and Minchin, R. F. (2000) Inactivation of human arylamine N-acetyltransferase 1 by the hydroxylamine of p-aminobenzoic acid. Biochem. Pharmacol. 60, 1829–1836. Hao, X-Y., Widersten, M., Ridderstro¨m, M., Hellman, U., and Mannervik, B. (1994) Co-variation of glutathione transferase expression and cytostatic drug resistance in HeLa cells: Establishment of class Mu glutathione transferase M3-3 as the dominating isoenzyme. Biochem. J. 297, 59–67. Lu, S. C., Sun, W.-M., Yi, J., Ookhtens, M., Sze, G., and Kaplowitz, N. (1996) Role of two recently cloned rat liver GSH transporters in
2016
(65)
(66) (67) (68)
(69)
(70)
(71)
(72) (73)
(74) (75) (76) (77)
Chem. Res. Toxicol., Vol. 21, No. 10, 2008
the ubiquitous transport of GSH in mammalian cells. J. Clin. InVest. 97, 1488–1496. Menon, S. G., Coleman, M. C., Walsh, S. A., Spitz, D. R., and Goswami, P. C. (2005) Differential susceptibility of nonmalignant human breast epithelial cells and breast cancer cells to thiol antioxidantinduced G1-delay. Antioxid. Redox Signaling 7, 711–718. Weber, W. W., and Hein, D. W. (1985) N-Acetylation pharmacogenetics. Pharmacol. ReV. 37, 26–79. Evans, D. A. P. (1989) N-Acetyltransferase. Pharmacol. Ther. 42, 157– 234. Atmane, N., Dairou, J., Paul, A., Dupret, J.-M., and Rodrigues-Lima, F. (2003) Redox regulation of the human xenobiotic metabolizing enzyme arylamine N-acetyltransferase 1 (NAT1). J. Biol. Chem. 278, 35086–35092. Dairou, J., Malecaze, F., Dupret, J.-M., and Rodrigues-Lima, F. (2005) The xenobiotic-metabolizing enzymes arylamine N-acetyltransferases in human lens epithelial cells: Inactivation by cellular oxidants and UVB-induced oxidative stress. Mol. Pharmacol. 67, 1299–1306. Dairou, J., Atmane, N., Rodrigues.Lima, F., and Dupret, J.-M. (2004) Peroxynitrite irreversibly inactivates the human xenobiotic-metabolizing enzyme arylamine N-acetyltransferase 1 (NAT1) in human breast cancer cells. J. Biol. Chem. 279, 7708–7714. Dairou, J., Dupret, J.-M., and Rodrigues-Lima, F. (2005) Impairment of the activity of the xenobiotic-metabolizing enzymes arylamine N-acetyltransferases 1 and 2 (NAT1/NAT2) by peroxynitrite in mouse skeletal muscle cells. FEBS Lett. 579, 4719–4723. King, R. S., and Duffel, M. W. (1997) Oxidation-dependent inactivation of aryl sulfotransferase IV by primary N-hydroxy arylamines during in vitro assays. Carcinogenesis 18, 843–849. Ragunathan, N., Dairou, J., Pluvinage, B., Martins, M., Petit, E., Janel, N., Dupret, J.-M., and Rodrigues-Lima, F. (2008) Identification of the xenobiotic-metabolizing enzyme arylamine N-acetyltransferase 1 (NAT1) as a new target of cisplatin in breast cancer cells: molecular and cellular mechanisms of inhibition. Mol. Pharmacol. 73, 1761– 1768. Szabo, C., Ischiropoulos, H., and Radi, R. (2007) Peroxynitrite: Biochemistry, pathophysiology and development of therapeutics. Nat. ReV. Drug DiscoVery 6, 662–680. Jatoe, S. D., Khan, S., and O’Brien, P. J. (1991) Molecular cytotoxic mechanism of carcinognic arylamines. Prog. Pharmacol. Clin. Pharmacol. 8, 245–253. Neumann, H.-G. (2007) Aromatic amines in experimental cancer research: Tissue-specific effects, an old problem and new solutions. Crit. ReV. Toxicol. 37, 211–236. Floyd, R. A., Soong, L. M., Stuart, M. A., and Reigh, D. L. (1978) Free radicals and carcinogenesis. Some properties of the nitroxyl free
Liu et al.
(78)
(79)
(80)
(81)
(82)
(83) (84) (85) (86)
(87)
radicals produced by covalent binding of 2-nitrosofluorene to unsaturated lipids of membranes. Arch. Biochem. Biophys. 185, 450–457. Naisbitt, D. J., Farrell, J., Gordon, S. F., Maggs, J. L., Burkhart, C., Pichler, W. J., Pirmohamed, M., and Park, B. K. (2002) Covalent binding of the nitroso metabolite of sulfamethoxazole leads to toxicity and major histocompatibility complex-restricted antigen presentation. Mol. Pharmacol. 62, 628–637. Vyas, P. M., Roychowdhury, S., Koukouritaki, S. B., Hines, R. N., Krueger, S. K., Williams, D. E., Nauseef, W. M., and Svensson, C. K. (2006) Enzyme-mediated protein haptenation of dapsone and sulfamethoxazole in human keratinocytes: II. Expression and role of flavin-containing monooxygenases and peroxidases. J. Pharmacol. Exp. Ther. 319, 497–505. Zhang, N., Liu, L., Liu, F., Wagner, C. R., Hanna, P. E., and Walters, K. J. (2006) NMR-based model reveals the structural determinants of mammalian arylamine N-acetyltransferase substrate specificity. J. Mol. Biol. 363, 188–200. Wu, H., Dombrovsky, L., Tempel, W., Martin, F., Loppnau, P., Goodfellow, G. H., Grant, D. M., and Plotnikov, A. N. (2007) Structural basis of substrate-binding specificity of human arylamine N-acetyltransferases. J. Biol. Chem. 282, 30189–30197. Wang, H., Vath, G. M., Gleason, K. J., Hanna, P. E., and Wagner, C. R. (2004) Probing the mechanism of hamster arylamine Nacetyltransferase 2 acetylation by active site modification, site-directed mutagenesis, and pre-steady state and steady state kinetic studies. Biochemistry 43, 8234–8246. Beije, B., and Moller, L. (1988) 2-Nitrofluorene and related compounds: Prevalence and biological effects. Mutat. Res. 196, 177–209. Zwirner-Baier, I., and Neumann, H.-G. (1999) Polycyclic nitroarenes (nitro-PAHs) as biomarkers of exposure to diesel exhaust. Mutat. Res. 441, 135–144. Boelsterli, U. A., Ho, H. K., Zhou, S., and Leow, K. Y. (2006) Bioactivation and hepatotoxicity of nitroaromatic drugs. Curr. Drug Metab. 7, 715–727. Adam, P. J., Berry, J., Loader, J. A., Tyson, K. L., Craggs, G., Smith, P., De.Belin, J., Steers, G., Pezzella, F., Sachsenmeir, K. F., Stamps, A. C., Herath, A., Sim, E., O’Hare, M. J., Harris, A. L., and Terrett, J. A. (2003) Arylamine N-acetyltransferase-1 is highly expressed in breast cancers and conveys enhanced growth and resistance to etoposide in vitro. Mol. Cancer Res. 1, 826–835. Sim, E., Pinter, K., Mushtaq, A., Upton, A., Sandy, J., Bhakta, S., and Noble, M. (2003) Arylamine N-acetyltransferases: A pharmacogenomic approach to drug metabolism and endogenous function. Biochem. Soc. Trans. 31, 615–619.
TX800215H