ArticlePDF Available

Nicotine inhibits the metabolic activation of the tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone in rats

Authors:

Abstract

The effect of nicotine on the metabolism of the tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) was studied in rats. [1-14C]NNK was s.c. injected at a dose of 0.08 mumol/kg. Co-administration of a 500-fold higher dose of nicotine (40 mumol/kg) did not reduce the overall urinary excretion of radioactivity. However, the metabolic pattern in 24 h urine was significantly changed. Metabolites resulting from NNK activation by alpha-hydroxylation were significantly (P < 0.001) reduced to 72% of the control. Detoxification to N-oxides and the glucuronide of 4-(methyl-nitrosamino)-1-(3-pyridyl)-1-butanol increased to 155% (P < 0.01) and 188% (P < 0.01) of the control respectively. These results suggest that nicotine, which occurs in concentrations up to 30,000-fold higher than NNK in mainstream smoke of cigarettes may have a protective effect against metabolic activation of NNK.
Carcinogenesls vol.15 no.5
pp.
1061-1064, 1994
Nicotine inhibits the metabolic activation of the tobacco-specific
nitrosamine 4-(methylnitrosamino)-l-(3-pyridyl)-l-butanone in rats+
E.Richter and A.R.Tricker1
Walther Straub-Insritut fflr Pharmakologie und Toxikologie, Ludwig-
Maximilians-UniversitSt MOnchen, Nussbaumstrasse 26, D-80336 Mflnchen
and 'Analytisch-biologisches Forschungslabor
Prof.
Dr med. Adlkofer,
Goethestrasse 20, D-80336 MQncben, Germany
The effect of
nicotine
on the metabolism of the tobacco-specific
nitrosamine 4-(methylnitrosamino)-l-(3-pyridyI)-l-butanone
(NNK) was studied in rats. [1-14C]NNK was s.c. injected at
a dose of 0.08 /unol/kg. Co-administration of a 500-fold higher
dose of nicotine (40 /unol/kg) did not reduce the overall
urinary excretion of radioactivity. However, the metabolic
pattern in 24 h urine was significantly changed. Metabolites
resulting from NNK activation by a-hydroxylation were
significantly (P <
0.001)
reduced to 72% of the control.
Detoxification to iV-oxides and the glucuronide of 4-(methyI-
nitrosamino)-l-(3-pyridyl)-l-butanol increased to 155%
(P < 0.01) and 188% (P < 0.01) of the control respectively.
These results suggest that nicotine, which occurs in concen-
trations up to 30 000-fold higher than NNK in mainstream
smoke of cigarettes may have a protective effect against
metabolic activation of NNK.
Introduction
The tobacco-specific nitrosamine 4-{methylnitrosamino)-l-
(3-pyridyl)-l-butanone (NNK*) has been suggested to be involved
in the induction of lung cancer in smokers (1,2). NNK is a po-
tent pulmonary carcinogen and also induces tumors of the nasal
mucosa, exocrine pancreas and liver in rats (1-3). Together with
A^-nitrosonornicotine (NNN), it may be involved in the etiology
of oral cancer in users of smokeless tobacco products (2).
NNK requires metabolic activation for expression of its
carcinogenicity. Studies in rodents conclusively demonstrate that
hydroxylation of the methylene and methyl carbons adjacent to
the A'-nitroso group (so-called a-hydroxylation) are the key
metabolic processes leading to the initiation of carcinogenesis
(1,4-9).
As shown in Figure
1
the common urinary metabolite
from these two reactions is 4-oxo-(3-pyridyl)butyric acid (keto
acid).
The product of NNK carbonyl reduction, 4-(methyl-
nitrosamino)-l-(3-pyridyl)-l-butanol (NNAL) is also a rodent
carcinogen (4,10). NNAL undergoes a-hydroxylation in a similar
way to NNK resulting in the formation of the common urinary
metabolite, 4-hydroxy-(3-pyridyl)butyric acid (hydroxy acid).
Detoxification pathways include glucuronidation of NNAL and
pyridine-A'-oxidation of both NNK and NNAL. All these
•Abbreviations: NNK, 4-(in(^lrdtiosarrdrio)-l-{3-pyrklyI)-l-butanone; NNN,
yV'-nitrosonomicotine; NNAL, 4-{mediylnitrosamirwH-(3-pyTidyI)-l-butanol;
hydroxy acid, 4-hydroxy-(3-pyridyl)butyric acid; keto acid, 4-oxo-(3-pyridyl)-
butyric acid; NNAL-GIuc, [4-{methylnitrosamino)-l-(3-pyridyl)-but-l-yl]-/3-
O-D-glucopyranosiduronic acid; NNAL-N-oxide, 4-{methylnitrosamino)-l-
(3-pyridyWV^Hide)-l-butanoJ; NNK-Moxide, 4^ir«hylnitrosamire>H-(3-pyridyl)-
JV-oxideH-butanone; HPB, 4-hydroxy-<3-pyridyI)-l-butanorje.
+Dedicated to Professor Dr R.Preussmarm on the occasion of his 65th birthday.
metabolic pathways have been demonstrated not only in rodent
species (1,4,10-13), but also in primates (14-16) and in human
tissues
(17
19).
In humans the presence of hemoglobin and DNA
adducts formed from NNK and/or NNN has been reported (20,
21).
Recendy, the NNK metabolites NNAL and its glucuronide
have been detected in 24 h urine of smokers (22).
The tobacco alkaloid nicotine, quantitatively the main component
of cigarette smoke, is present at 3000- to 30 000-fold higher
concentrations than NNK in the mainstream and sidestream
smoke (23,24). In rat oral tissue, nicotine inhibits the metabolic
activation of NNK when present at 100-fold higher concentrations
dian NNK (25). For the rabbit olfactory-specific cytochrome
P450 isozyme NMa, nicotine is a competitive inhibitor of NNK
a-hydroxylation (26). The only in vivo study using a NNK dose
far in excess of that of
nicotine
did not show any effect of nicotine
on NNK activation
(27).
Therefore, we investigated the metabolism
of NNK in the rat when given togedier widi a 500-fold higher
dose of nicotine.
Materials and methods
Chemicals
[1-MC]NNK with a sp. act. of 29 mCi/mmol was obtained from Chemsyn
Science Laboratories (Lenexa, KS). NNK metabolite standards were a gift from Dr
D.Hoffmann (American Health Foundation, Valhalla, NY). (-)Nicotine,
0-glucuronidase (type DC) and thimerosal (sodium ethylmercurithiosalicylate)
were obtained from Sigma Chemie GmbH (Tauflrirchen, Germany). All other
chemicals, which were of either HPLC or analytical grade, were purchased from
Merck (Darmstadt, Germany).
Animals
Male Wistar rats (85-110 g) from the breeding colony of the University of
Gomngen were housed in stainless sted metabolism cages in a fully air-conditioned
room (18 ± 1°C; 60 ± 5% humidity). The day-night cycle was 12 h (light
from 7 a.m.). The animals had unrestricted access to Alma H 1003 Laboratory
chow (F.Botzenhardt KG, Kempten, Germany) and drinking water. The animal
experiments were officially approved by the Government of Upper Bavaria
(AZ 211-2531-53/92).
Collection of rat urine
Groups of eight rats were administered 0.4 ml of saline with either
[
1-14C]NNK
(8 nmol = 1.66 pg) alone or [1-14C]NNK plus nicotine (4 ^mol = 650 /ig) by
s.c. injection. Urine was collected over 24 h time intervals for 2 days in
polyethylene vials containing a few grains of thimerosal. Aliquots of 100 and
500 itl from the first and second day of the experiment respectively were used
for total 14C determination. The remaining urine was cenlrifuged and the
supernatant stored at -20°C until analysis by HPLC.
HPLC analysis
Urine samples from the first 24 h of the experiment were chromatographed on
a 4.6 x 250 mm UChrosorb* RP18 SelectB column (Merck) by elution with
a gradient of 100% A for 0.5 min, linear to 80% A/20% B in 20 min and linear
to 20% A/80% B in 2 min (A: 20 mM Tris buffer, pH 7.2; B: acetonkrile) at
a flow rate of 0.7 ml/min. Detection of 14C was performed by solid-phase
radioactivity monitoring (Ramona, Raytest, Straubenhardt, Germany). Radioactive
metabolites were identified by co-chromatography with unlabeled reference
compounds detected by UV at 234 and 254 nm (UVD 160, Gynkotek, Germering
Germany). The O-ghicuronide of NNAL was further characterized by co-injecting
the purified radioactive compound obtained in a previous study (12) and by
treatment of urine with 0-glucuronidase (13).
Suaistical analysis
Reported values represent means ± standard error. Statistical analysis was
performed by the two-sided Mest for independent samples.
© Oxford University Press1061
E.Rkhter and A.R.Tricker
>T NNAL-Qfcic
HPBhy*cny»dd
Fig. 1. Metabolic scheme of NNK. Structures in brackets are hypothetical intermediates (13).
Results
The urinary excretion of NNK was studied in rats s.c. injected
with ~ 80 nmol/kg NNK either alone or in combination with
40 /imol/kg nicotine. This represents a 500-fold higher dose
of nicotine than NNK. In excess of 98% of the total urinary
excretion occurred within the first 24 h with no difference
between NNK only (72.4 ± 2.9%) and NNK plus nicotine
(74.8 ± 2.1%) treated rats.
Figure 2 shows typical HPLC profiles of
the
urinary metabolites
of [1-I4C]NNK obtained from rats treated with or without a
500-fold excess of
nicotine.
Peaks I and II co-eluted with hydroxy
acid and 4-oxo-4-(3-pyridyl)butyric acid (keto acid) resulting from
a-hydroxylation of NNK and NNAL respectively (Figure 1).
Peak in corresponds to [4-(methylnitrosamino)-l-(3-pyridyl)-
but-l-yl]-/3-OD-glucopyranosiduronic acid (NNAL-Gluc). Peaks
IV and V co-eluted with 4-(methylnitrosamino)-l-(3-pyridyl-
Ak)xide)-l-butanol (NNAL-A^-oxide) and 4-<methylnitrosamino)-
l-(3-pyridyl-A^-oxide)-l-butanone (NNK-N-oxide). 4-Hydroxy-
l-(3-pyridyl)-butanol (diol), 4-oxo-l-(3-pyridyl)-l-butanone (HPB)
and NNAL were occasionally detected. Of
these
three metabolites,
only NNAL exceeded 2% of the radioactivity in five and two
of eight samples of 24 h urine of the NNK only and NNK plus
nicotine-treated rats respectively.
Figure 3 shows the pattern of
the
five major NNK metabolites
in 24 h urine. Nicotine treatment significantly reduced the
formation of hydroxy acid and keto acid to
75%
(P < 0.001) and
69%
(P < 0.001) of
the
control respectively. The detoxification
products NNAL-Gluc and NNAL-N-oxide were significantly
increased by 188% (P < 0.01) and 163% (P < 0.001) of the
control
respectively
by co-administration of
nicotine.
The formation
of NNK-N-oxide was also increased 124% but the difference did
not reach statistical significance. In Table I the results are
presented as total amounts excreted in 24 h urine.
Discussion
Cigarette smoke contains several thousand different components,
of which nicotine is quantitatively the most abundant. Nicotine
occurs in concentrations 3000- to 30 000-fold higher than NNK
in mainstream cigarette smoke (23,24). As such, nicotine was
chosen as the first candidate with which to study the in vivo
inhibition of NNK metabolism. Previous in vitro studies have
shown that nicotine is a potent competitive inhibitor of NNK
a-hydroxylation in hamster lung (28), rat oral tissue (25) and
in rabbit nasal olfactory and respiratory microsomes (26). The
effect of multiple-dose exposure to nicotine on the in vivo
metabolism of NNK in rats (27) and in vitro pulmonary
metabolism of NNK in hamsters (29) has also been studied. In
both studies, 0.002% nicotine was administered in drinking water
for 14 days. After nicotine pretreatment, no inhibition of
in
vivo
metabolic activation of NNK was observed in rats given a single
i.v. dose of 0.4 mmol NNK/kg body wt (27). Contrary to this
finding, nicotine pretreatment of hamsters induced pulmonary
a-hydroxylation in lung explants (29). In both studies, the nicotine
dose was 2000- to 20 000-fold lower than that of NNK.
In the reported study, the nicotine dose was 500-fold higher
than that of NNK. Subcutaneous injection of both nicotine and
NNK was chosen as the route of administration. Following s.c.
injection, NNK is first transported to the lung, which is considered
to be the primary site of NNK metabolism. Previous studies using
N-nitrosodibutylamine have shown a high first-pass metabolism
in the lung following either s.c. or i.v. administration (30).
Numerous studies with isolated perfused rat lung (31) as well
as different in
vitro
preparations of rat lung (5,9,11,32—36) have
demonstrated extensive metabolism of NNK by a-hydroxylation,
N-oxidation and reduction to NNAL in this organ. Quantitative
comparison of urinary NNK metabolites in Wistar rats clearly
shows nicotine inhibition of the metabolic activation of this
1062
Nicotine Inhibition of NNK metabolism
21.0
15.0
II. 1
5.0
I.I
CPS
i i i i
c
iiNNK control
IV V
vwJW-WlVw*\v
1UTable I. Effect of nicotine on excretion of NNK metabolites in 24 h urine1
21
15
11
5
1
I
I
I
1
-
CPS
1 1 1 1—I 1 T i i
NNK
1
+ nicotine
L
i
i i i
HC
1
III
15.11
Time (min)
Fig. 2. HPLC analysis of metabolites in the urine of rats 24 h following s.c.
administration of [1-14C]NNK at 80 nmol/kg with or without nicotine at
40 /tmol/kg. Labeling of radioactive peaks—hydroxy acid (I), keto acid (Tf),
NNAL-Gluc (III), NNAL-W-oxide (IV) and NNK-Ak>xide (V)—refers to
peaks co-eluting wim authentic standards added as UV markers.
o 40-
t
"S
30-
M-
o
^ 20-
c
IV
o
a 10-
f
1
T m
1
I
1 1
NNK control
NNK + nicotine
**
1
is A
III
Hydroxy
acidKeto
acid
NNALglu-
NNAL NNK
curonide N-oxide N-oxide
F^.
3. Effect of nicotine on die metabolite pattern in 24 h urine following
s.c. administration of [1-MC]NNK at 80 nmol/kg with or without nicotine at
40 /tmol/kg. Values are die mean ± SE of eight samples. Asterisks indicate
a statistically significant difference to the group given only NNK at
*P < 0.01 and **P <
0.001.
tobacco-specific nitrosamine (Table I). The largest suppression
was observed for the keto acid, which results from a-hydroxylation
of NNK (Figure 1). This decrease was not compensated by
Metabolitepmol detected in urine (% of urinary metabolites)
Hydroxy acid
Keto acid
NNAL-Gluc
NNAL-A'-oxide
NNK-Moxide
£ of alphab
£ of N-oxxtes
NNAL-Gluc + NNALC
Total
NNK
668 db 17
1332 ± 50
118 ± 20
441 ± 38
320 ± 37
2021 ± 57
720 ± 79
161 ± 21
2902 ±104
(22.4)d
(44.7)
(3.9)
(13.3)
(10.6)
(67.8)
(23.8)
(6.2)
(96.9)
NNK
502
919
222
719
397
1462
1116
296
2874
+
±
±
±
±
±
±
±
nicotine
24
35
26
27
54
41
68
41
105
(17.6)***
(31.9)***
(7.6)**
(23.0)***
(13.5)
(50.8)***
(38.5)**
(10.1)*
(99.3)
•Groups of eight male Wistar rats were administered [1-14C]NNK at a dose
of 80 nmol/kg wim or wimout nicotine at a dose of 40 /imol/kg. Urine was
collected for 24 h and analyzed for urinary metabolites of NNK as indicated
in the text.
'including hydroxy acid, keto acid, diol (detected in one sample from each
experiment) and HPB (detected in four and five samples from experiments
wim NNK and NNK + nicotine respectively).
"^NNAL was detected in five and six samples from experiments with NNK
and NNK + nicotine respectively.
dMean ± SE of eight samples. Values labeled are statistically significandy
different from the NNK group at *P < 0.05, **P < 0.01 and
••*/> <
0.001.
a comparable increase in NNK-ALoxide formation. Therefore,
the in vivo equilibrium existing between NNK and NNAL
as described by Adams et al. (15) is shifted in favor of
NNAL. The urinary excretion of hydroxy acid resulting from
a-hydroxylation of NNAL was also reduced. The excretion
of NNAL and its detoxification products, NNAL-Gluc and
NNAL-A'-oxide, expressed as total urinary excretion of I4C
nearly doubled from 19.9 ± 1.5% in control rats to
35.1 ± 1.0% in rats treated with nicotine.
Whether nicotine specifically inhibits metabolic activation of
NNK by a-hydroxylation at either the methylene or methyl
carbon atom adjacent to the N-nitroso group or both sites
simultaneously, cannot be determined from the present results.
Methylene hydroxylation produces methanediazohydroxide,
which can methylate DNA bases in vivo. Methyl hydroxylation
yields 4-(3-pyridyl)-4-oxobutanediazohydroxide, which pyridyl-
oxobutylates DNA. Which of these two pathways plays the
predominant role in the proposed carcinogenic effect of NNK
in humans is unknown (37). The extent of pyridyloxobutylation
of DNA by NNK can be estimated by measuring HPB released
by alkaline hydrolysis of hemoglobin (38). This HPB-releasing
adduct is also formed by NNN (39) and has been quantified in
tobacco users, since it is considered to be a surrogate marker
for the uptake and activation of both NNN and NNK (20). Such
studies show a < 3-fold difference in HPB-releasing hemoglobin
adduct levels in smokers and nonsmokers (20,40,41). The results
of the present study clearly show that nicotine inhibits NNK
a-hydroxylation, the required metabolic step in the formation
of HPB-releasing adducts.
Additional comparative studies need to be performed with
NNN, both NNN and NNK in combination and with a higher
nicotine dose relative to both nitrosamines to determine the full
extent of nicotine inhibition of tobacco-specific nitrosamine
metabolism. Furthermore, the mechanisms of nicotine inhibition
of NNK metabolism and both hemoglobin and tissue adduct
formation need to be studied.
1063
E.RJchter and A.R.Tricker
Acknowledgements
We extend special thanks to D.Hoffmann for providing reference compounds
and Christiana Oehlmann for expert technical assistance. This work was supported
by a grant from the Forschungsrat Rauchen und Gesundheh.
References
1.
Hecht.S.S. and Hoffmann.D. (1988) Tobacco-specifk njtrosamines, an
important group of carcinogens in tobacco and tobacco smoke.
Carcinogenesis,
9, 875-884.
2.
Hecht.S.S. and Hoffmann.D. (1989) The relevance of tobacco-specific
nitrosamines to human cancer. Cancer Surv., 8, 273—294.
3.
Rivenson.A., Hoffmann,D., Prokopczyk,B., Amin.S. and Hecht.S.S. (1988)
Induction of lung and exocrine pancreas tumors in F344 rats by tobacco-specific
and Areca-dcrivcd Mnhrosamines. Cancer Res., 48, 6912-6917.
4.
Castonguay.A., Tjalve.H. and Hecht.S.S. (1983) Tissue distribution of the
tobacco-specific carcinogen 4-(memylnitrosamino)-l-(3-pyridyl)-l-butanone
and its metabolites in F344 rats. Cancer Res., 43, 630-638.
5.
Bdinsky.S.A., FoteyJ.F., White.C.M., Anderson.M.W. and Maronpot, R.R.
(1990) Dose—response relationship between C^-methylguanine formation in
Clara cells and induction of pulmonary neoplasia in the rat by
4-(nWrrylnitrc)samino)-l-(3-pyridyI)-l-butanone. CancerRes., 50, 3772-3780.
6. Hecht.S.S., Trushin.N., Castonguay.A. and Rivenson,A. (1986) Comparative
tumorigenicity and DNA methylation in F344 rats by 4-(methylnitrosamino)-
l-(3-pyridyl)-l-butanone and A^-nitrosodimethylamine. Cancer Res., 46,
489-502.
7.
Hecht.S.S., Spratt.T.E. and Trushin.N. (1988) Evidence for 4-(3-pyridyl)-
4-oxobutylatk>n of DNA in F344 rats treated with the tobacco-specific
nitrosamines 4-(methylnitrosamino)-l-(3-pyridyl)-l-butanone and A^nhroso-
nomicotine. Carcinogenesis, 9,
161
165.
8. Peterson.L.A. and Hecht.S.S. (1991) O*-Methylguanine is a critical
determinant of 4-(memylnitrosamino)-l-(3-pyridyl)-l-butanone tumorigenesis
in A/J mouse lung. Cancer Res., 51, 5557-5564.
9. Belinsky.S.A., Devereux.T.R., White,C.M., FoleyJ.F., Maronpot.R.R. and
Anderson.M.W. (1991) Role of Clara cells and type n cells in the development
of pulmonary tumors in rats and mice following exposure to a tobacco-specific
nitrosamine. Exp. Lung Res., 17, 263-278.
10.
Castonguay.A., Lin.D., Stoner.G.D., Radok.P., Furuya.K., Hecht.S.S.,
Schut.H.A.J. and Klaunig,J.E. (1983) Comparative carcinogenicity in A/J
mice and metabolism by cultured mouse peripheral lung of A^-mtrosonornicotine,
4-(memylnhTosamino)-l-(3-pyridyl)-l-butanone, and their analogues. Cancer
Res.,
43, 1223-1229.
11.
Bdinsky.S.A., White.C.M., Trushin.N. and Hecht.S.S. (1989) Cell specificity
for die pulmonary metabolism of tobacco-specific nitrosamines in the Fischer
rat. Carcinogenesis, 10, 2269-2274.
12.
SchulzeJ., Richter.E., Binder.U. and Zwickenpflug.W. (1992) Biliary
excretion of 4-(methylnitrosamino)-l-(3-pyridyl)-l-butanone in die rat.
Carcinogenesis, 13, 1961-1965.
13.
Morse.M.A., Eldind^C.I., Toussaint.M., Amin.S.G. andChung,F.-L. (1990)
Characterization of a glucuronide metabolite of 4-(methylrutrosamino)-
l-(3-pyridyl)-l-butanone (NNK) and its dose-dependent excretion in the urine
of mice and rats. Carcinogenesis, 11, 1819-1823.
14.
Castonguay.A., Tjalve.H., Trushin.N., D'Argy.R. and Sperber.G. (1985)
Metabolism and tissue distribution of tobacco-specific nitrosamines in die
marmoset monkey
(CaUithrix
jacchus). Carcinogenesis, 6, 1543—1550.
15.
AdamsJ.D., Lavoie.EJ., O'Mara-Adams.KJ., Hoffinann.D., Carey.K.D.
and Marshall.M.V. (1985) Pharmacolrinetics of A^-nitrosonornicotine and
4-(methylnitrosaniino)-l-(3-pyridyl)-l-butanone in laboratory animals. Cancer
Lett., 28,
195-201.
16.
Hecht,S.S., Trushin,N., Reid-Quinn,C.A., Burak,E.S., Jones,A.B., Southers,
J.L., Gombar.C.T., Carmella.S.G., Anderson.L.M. and RiceJ.M. (1993)
Metabolism of the tobacco-specific nitrosamine 4-(methylnitrosamino)-
l-(3-pyridyl)-l-butanone in the patas monkey: pharmacokinetics and
characterization of glucuronide metabolites. Carcinogenesis, 14, 229-236.
17.
CastonguayA, Stoner.G.D., Schut,H.AJ. and Hecht,S.S. (1983) Metabolism
of tobacco-specific A'-nitrosammes by cultured human tissues. Proc. NatL
Acad.
Sd. USA, 80, 6694-6697.
18.
Smith.T.J., Guo.Z., Gonzalez.F.J., Guengerich.F.P., Stoner.G.D. and
Yang.C.S.
(1992) Metabolism of 4-(methyuiitrosamino)-l-(3-pyridyl)-
1-butanone
in human lung and liver microsomes and cytochromes P-450
expressed in hepatoma cells. CancerRes., 52, 1757-1763.
19.
Yamazaki.H., Inui.Y., Yun,C.-H., Guengerich.F.P. and Shimada.T. (1992)
Cytochrome P450 2E1 and 2A6 enzymes as major catalysts for metabolic
activation of ALaitrosodialkylamines and tobacco-related nitrosamines in human
liver microsomes. Carcinogenesis, 13, 1789-1794.
20.
Carmella,S.G., Kagan.S.S., Kagan.M., Foiles.P.G., Palladino.G.,
Quart.A.M., Quart.E. and Hecht.S.S. (1990) Mass spectrometric analysis
of tobacco-specific nitrosamine hemoglobin adducts in snuff-dippers, smokers,
and non-smokers. Cancer Res., 50, 5438-5445.
21.
Foiles.P.G., Akerkar.S.A., Carmella.S.G., Kagan.M., Stoner.G.D.,
ResauJ.H. and Hecht.S.S. (1991) Mass spectrometric analysis of tobacco-
specific nitrosamine DNA adducts in smokers and nonsmokers. Chem. Res.
Toxicol., 4, 364-368.
22.
Carmella.S.G., Akerkar.S. and Hecht.S.S. (1993) Metabolites of
the
tobacco-
specific nitrosamine 4-{methylnitrosamino)-l-(3-pyridyl)-l-butanone in
smokers' urine. CancerRes., 53, 721-724.
23.
AdamsJ.D., O'Mara-Adams.K.J. and Hoffmann.D. (1987) Toxic and
carcinogenic agents in undiluted mainstream smoke and sidestream smoke
of different types of cigarettes. Carcinogenesis, 8,
729—731.
24.
Tricker.A.R., Ditrich.C. and Preussmann.R. (1991) W-Nitroso compounds
in cigarette tobacco and their occurrence in mainstream tobacco smoke.
Carcinogenesis, 12,
257-261.
25.
Murphy,S.E. and Heiblum.R. (1990) Effect of nicotine and tobacco-specific
nitrosamines on the metabolism of A^-nitrosonomicotine and 4-(methyl-
nitrosamino)-l-(3-pyridyl)-l-butanone by rat oral tissue.
Carcinogenesis,
11,
1663-1666.
26.
HongJ.-Y., Ding.X., Smith.T.J., Coon.M.J. and Yang.C.S. (1992)
Metabolism of 4-(methylnitrosamino)-l-(3-pyridyl)-l-butanone (NNK), a
tobacco-specific carcinogen, by rabbit nasal microsomes and cytochrome P450s
NMa and NMb. Carcinogenesis, 13, 2141-2144.
27.
Prokopczyk.G., Adams,J.D., LeVoie.E.J. and Hoffmann.D. (1987) Effect
of snuff and nicotine on DNA methylation and 4-(methylnitrosamino>-
K3-pyrklyl)-l-butanone. Carcinogenesis, 8, 1395-1397.
28.
Schuller.H.M., Castonguay.A., Orloff.M. and Rossignol.G. (1991)
Modulation of the uptake and metabolism of 4-(methylnitrosamino)-
l-(3-pyridyl)-l-butanone by nicotine in hamster lung. Cancer Res., 51,
2009-2014.
29.
Charest.M., Rossignol.G. and Castonguay.A. (1989) In vitro and in vivo
modulation of die bioactivation of 4-(methylnitrosamino)-l-(3-pyridyl)-
1-butanone
in hamster lung tissues. Chem.-Biol. Interactions, 71, 265 -278.
30.
Feng,X.-C. and Richter.E. (1989) The role of extrahepatic organs in the first
pass metabolism of A'-nitrosodibutylamine. Arch. Toxicol. Suppl., 13,
227-229.
31.
Foth.H., Schulze.J. and Richter.E. (1991) NNK metabolism by isolated
perfused rat lung. IARC Tech. Report, 11, P29.
32.
Doerr-O'Rourke.K., Trushin.N., Hecht.S.S. and Stoner.G.D. (1991) Effect
of phenethyl isothiocyanate on the metabolism of die tobacco-specific
nitrosamine 4-(methylnitrosamino)-l-(3-pyridyl)-l-butanone by cultured rat
lung tissue. Carcinogenesis, 12,
1029
1034.
33.
Guo.Z., Smith.T.J., Thomas.P.E. and Yang.C.S. (1991) Metabolic activation
of 4-(methylnitrosamino)-l-(3-pyridyl)-l-butanone as measured by DNA
alkylation in vitro and its inhibition by isothiocyanates. Cancer Res., 51,
4798-4803.
34.
Peterson.L.A., Mathew.R. and Hecht.S.S. (1991) Quantitation of microsomal
alpha-hydroxylation of the tobacco-specific nitrosamine, 4-(methylnitrosamino)-
l-(3-pyridyl)-l-butanone. CancerRes., 51, 5495-5500.
35.
Peterson.L.A., Mathew.R., Murphy.S.E., Trushin.N. and Hecht.S.S. (1991)
In vivo and in vitro persistence of pyridyloxobutyl DNA adducts from
4-(methylnitrosamino)-l-(3-pyridyl)-l-butanone. Carcinogenesis, 12,
2069-2072.
36.
Guo.Z.Y., Smith.TJ., Thomas.P.E. and Yang.C.S. (1992) Metabolism of
4-(rriethylnitrosarnino)-l-{3-pyridyl)-l-butanone by inducible and constitutive
cytochrome P450 enzymes in rats. Arch. Biochem. Biophys., 298, 279-286.
37.
Hecht.S.S., Lin.D., Castonguay.A. and Rivenson.A. (1987) Effects of
a-deuterium substitution on die tumorigenicity of 4-(methylnitrosamino)-
l-<3-pyridyi)-l-butanone in F344 rats. Carcinogenesis, 8, 291-294.
38.
Murphy.S.E., Paloraino.A., Hecht.S.S. and Hoffmann.D. (1990)
Dose—response study of DNA and hemoglobin adduct formation by
4-(methylnitrosamino)-l-(3-pyridyl)-l-butanone in F344 rats. Cancer Res.,
50,
5446-5452.
39.
Carmella,S.G. and Hecht,S.S. (1987) Formation of hemoglobin adducts upon
treatment of F344 rats with the tobacco-specific nitrosamines 4-(methyl-
nitrosamino)-l-(3-pyridyI)-l-butanone and Ar-nilrosonomicotine.
Cancer
Res.,
47,
2626-2630.
40.
Falter,B., Kutzer.C. and Richter.E. (1994) Bkmxmitoring of hemoglobin
adducts: aromatic amines and tobacco-specific nitrosamines. CUn. Invest.,
in press.
41.
Richter.E., Schaffler.G., Malone.A. and SchulzeJ. (1992) Tobacco-specific
nitrosamines—metabolism and biological monitoring of exposure to tobacco
products. CUn. Invest., 70, 290-294.
Received on
July
6,
1993;
revised
on
December
15, 1993;
accepted
on
December
23,
1993
1064
... Exposure to nicotine caused a statistically significant increase in micronucleus frequency in human gingival fibroblasts in vitro [25] . However, it has been demonstrated that nicotine inhibits the action of nitrosamines which is catalyzed by P450 2E1 [26,27] , and also it participates in the conversion of benzo[a]pyrene to DNA-reactive metabolites [28] . Nitrosamines and polycyclic aromatic amines are considered important chemical agents able to promote genetic insult as far as carcinogenesis [15] . ...
Article
Objective: Nowadays, much attention has been focused on the search for new non-invasive methodologies able to predict malignant transformation of oral mucosa cells. The aim of the present study was to comparatively evaluate DNA damage (micronucleus) and cellular death (pyknosis, karyolysis and karyorrhexis) in exfoliated oral mucosa cells from smokers and non-smokers in buccal mucosa cells. Study design: A total of 24 young, healthy smokers and 14 non-smokers were included in this setting. Individuals had epithelial cells from the cheek mechanically exfoliated, placed in fixative and dropped in clean slides which were checked for the above nuclear phenotypes. Results: Smokers presented more (p < 0.05) micronucleated oral mucosa cells than non-smokers. Tobacco smoke was not able to increase other nuclear alterations closely related to cytotoxicity such as karyorrhexis, pyknosis and karyolysis. Conclusion: In summary, these data indicate that the cigarette is able to induce micronuclei in oral mucosa cells, so the micronucleus test is a suitable method for predicting oral cancer risk.
Chapter
The pathogenesis of the major smoking-related diseases, cancer of the lung and coronary heart disease, are poorly understood. There are, however, well founded hypotheses how smoking could contribute to the development of these diseases. On the other hand, the constituents of cigarette smoke that might be the causative agents remain to be identified. At present, there is no evidence at all that nicotine — the substance most smokers smoke for — has any influence on the development of cancer in otherwise healthy smokers, and only spurious evidence that it may be involved in the development of coronary heart disease.
Chapter
Full-text available
[5- 3H]-4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) was infused subcutaneously at a daily dose of 7.2 nmol/kg with or without 26 µmol/kg nicotine or cotinine, respectively, over four weeks in male F344 rats. Coadministration of nicotine or cotinine had no effect on the excretion of NNK and its metabolites. However, at the end of the experiment binding of radioactivity to hemoglobin in nicotine- and cotinine-treated rats was reduced by 50% compared to NNK-only treated rats. The inhibition of hemoglobin adduct formation by tobacco alkaloids may explain the lower than expected differences in the adduct levels of tobacco-specific nitrosamines between smokers and nonsmokers as observed in several biomonitoring studies.
Chapter
The secretion of several growth factors by pulmonary neuro-endocrine (PNE) cells is under cholinergic control, and nicotine stimulates the proliferation of fetal PNE cells in vivo by binding to the nicotinic acetylcholine receptor (nAChR). Our data show that fetal hamster PNE cells and cell lines derived from human neuroendocrine lung cancers require simultaneous stimulation of the oxygen receptor and the nAChR expressed in these cells in order to proliferate in response to nicotine. The simultaneous stimulation of these two receptor-initiated signal transduction pathways triggers secretion of 5-HT and bombesin, and activates protein kinase C (PKC) and c-fos downstream. The importance of this synergism is greatly emphasized by our finding that simultaneous exposure of hamsters to hyperoxia and nicotine causes lung tumors positive for 5-HT and neuron specific enolase (NSE) while nicotine alone or hyperoxia alone cause no tumors.
Chapter
While nicotine is not a carcinogen, several tobacco-specific nitrosamines derived from nicotine and other tobacco alkaloids are carcinogenic in laboratory animals; a property characteristic of over 200 nitrosamines 4-(methylnitrosamino)-l-(3-pyridyl)-l-butanone (NNK), 4-(methylnitrosamino)-l-(3-pyridyl)- 1-butanol (NNAL), and N'-nitrosonornicotine (NNN) are strong rodent carcinogens, while N'-Nitrosoanabasine (NAB), N'-nitrosoanatabine (NAT), iso-NNAL, and 4-(methylnitrosamino)- l-(3-pyridyl)butyric acid (iso-NNAC) have little or no activity. The carcinogenicity of NNK, NNAL, and NNN leads to the hypothesis that they may play an important role in human cancer. In support of this hypothesis, numerous analytical studies summarized the presence of tobacco-specific nitrosamines in cured, unburned tobacco, as well as in tobacco smoke. Virtually, all marketed tobacco products contain these compounds. Numerous studies in rodents and primates, both in vitro and in vivo, demonstrate that NNK, NNAL, and NNN are extensively metabolized and form electrophilic intermediates that form covalent adducts with DNA and hemoglobin. These studies provide the mechanistic foundation for understanding the carcinogenic activities of tobacco-specific nitrosamines. The results of the carcinogenicity studies of NNK, NNAL, and NNN further support the hypothesis that these nitrosamines may be important in tobacco induced cancer. The structural similarities of NNK, NNAL, and NNN to nicotine indicate that these nitrosamines, like nicotine, should be extensively metabolized in humans; this has been difficult to demonstrate so far, due in part to the identical structures of nicotine and tobacco-specific nitrosamine metabolites.
Article
Few clinical studies involving cigarettes have provided a comprehensive picture of smoke exposure, test article characterization, and insights into sensory properties combined. The purpose of these pilot studies was to determine whether cigarettes with different levels of nicotine but similar tar levels would affect sensory experience or smoking behavior so as to significantly alter levels of selected biomarkers of exposure (BOE). In 2 confined, double-blind studies, 120 adult smokers switched from Marlboro Gold cigarettes at baseline to either 1 of 2 lower nicotine cigarettes or 1 of 2 higher nicotine cigarettes and then to the other cigarette after 5 days. Urinary excretion of exposure biomarkers (nicotine equivalents [NE], total and free 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol [NNAL], 1-hydroxypyrene, and 3-hydroxypropyl mercapturic acid) as well as carboxyhemoglobin and plasma cotinine were measured at baseline, Day 5, and Day 10. Daily cigarette consumption was monitored and sensory characteristics were rated for each cigarette. With higher nicotine yield, urine NE, urine total NNAL, and plasma cotinine increased, while nonnicotine BOE decreased, without changes in cigarette consumption. In contrast, with lower nicotine yield, urine NE, urine total NNAL, and plasma cotinine dropped, while nonnicotine BOE and cigarettes per day increased. Higher nicotine cigarettes were rated "harsher" and "stronger" than baseline, while lower nicotine cigarettes were less "strong." All 4 test cigarettes were "highly disliked." These studies demonstrated that abrupt increases or decreases in nicotine and the resulting sensory changes impact BOE through changes in intensity or frequency of smoking.
Article
In non-smokers, haemoglobin adducts from 3- and 4-aminobiphenyl have been reported to arise mainly from exposure to environmental tobacco smoke (ETS). Therefore, the impact of self-reported smoking (n = 27) and exposure of non-smokers to ETS (n = 78) on haemoglobin adducts was studied in pregnant women from Homburg, Germany. In addition to 3- and 4-aminobiphenyl, adducts from seven monocyclic aromatic amines (aniline, o -, m -, and p -toluidine, 2,4-dimethylaniline, 2-ethylaniline and o -anisidine) and the adduct from tobacco-specific nitrosamines (4-hydroxy-1-(3-pyridyl)1-butanone) were determined. Five of 78 self-reported non-smoking women had plasma cotinine levels and urinary cotinine/creatinine ratios indicative of active smoking. In the remaining 73 non-smokers cotinine/creatinine ratios correlated significantly with self reported exposure to ETS. However, none of the haemoglobin adducts increased with increasing exposure to ETS or increasing cotinine/creatinine ratios. Although significantly elevated in smoking compared with non-smoking women, the mean haemoglobin adduct levels formed by tobacco-specific nitrosamines (54 7 8 9 vs 26 7 4 1 fmol g-1, p < 0 001), 3-aminobiphenyl (3 0 0 5 vs 1 4 0 1 pg g-1, p < 0 001), 4-aminobiphenyl (27 9 3 4 vs 10 2 0 7 pg g-1, p < 0 001), o -toluidine (289 25 vs 237 65 pg g-1, p < 0 001), p -toluidine (315 32 vs 197 13 pg g-1; p < 0 001), 2,4-dimethylaniline (25 5 2 9 vs 18 6 1 6 pg g-1, p < 0 05), had considerable overlappings ranges indicating lack of specificity as biomarkers to tobacco smoke exposure. Exposure to other as yet unknown environmental sources appearsto be more significant than previously thought.
Article
The exposure of non-smokers to the tobacco-specific N-nitrosamine 4-(N-methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), a rodent lung carcinogen, was determined in the air of various indoor environments as well as by biomonitoring of non-smokers exposed to environmental tobacco smoke (ETS) under real-life conditions using the urinary NNK metabolites 4-(N-methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL) and [4-(N-methylnitrosamino)-1-(3-pyridyl)but-1-yl]-beta-O-D-glucosiduronic acid (NNAL-Gluc). NNK was not detectable (<0.5 ng m(-3)) in 11 rooms in which smoking did not occur. The mean NNK concentration in 19 rooms in which smoking took place was 17.5 (2.4-50.0) ng m(-3). The NNK levels significantly correlated with the nicotine levels (r=0.856; p< 0.0001). Of the 29 non-smokers investigated, 12 exhibited no detectable NNAL and NNAL-Gluc excretion (<3 pmol day) in their urine. The mean urinary excretion of NNAL and NNAL-Gluc of the 17 remaining non-smokers was 20.3 (<3-63.2) and 22.9 (<3-90.0) pmol day(-1), respectively. Total NNAL excretion (NNAL+NNAL-Gluc) in all non-smokers investigated significantly correlated with the amount of nicotine on personal samplers worn during the week prior to urine collection (r=0.88; <0.0001) and with the urinary cotinine levels (r=0.40; p=0.038). No correlation was found between NNAL excretion and the reported extent of ETS exposure. Average total NNAL excretion in the non-smokers with detectable NNAL levels was 74 times less than in 20 smokers who were also investigated. The cotinine/total NNAL ratios in urine of smokers (9900) and non-smokers (9300) were similar. This appears to be at variance with the ratios of the corresponding precursors (nicotine/NNK) in mainstream smoke (16400) and ETS (1000). Possible reasons for this discrepancy are discussed. The possible role of NNK as a lung carcinogen in non-smokers is unclear, especially since NNK exposure in non-smokers is several orders of magnitude lower than the ordinary exposure to exogenous and endogenous N-nitrosamines and the role of NNK as a human lung carcinogen is not fully understood.
Article
Background: Tobacco-specific nitrosamines (TSNAs) are a group of carcinogens, which originate from nicotine and other tobacco alkaloids during fermentation and burning of tobacco. Between 1990 and 2010, the tobacco industry-funded extensive academic research on TSNAs in Germany. The objective was to gain better knowledge of how industry aims and strategies correlate with contents of publications by German toxicologists accepting tobacco industry funding by focusing on one prominent such toxicologist. Methods and findings: The authors analysed previously secret tobacco industry documents that were disclosed following a series of litigation cases in the USA and compared them with peer-reviewed published results of tobacco industry-funded toxicologists. The tobacco industry, in particular Philip Morris, developed sophisticated strategies to downplay TSNA's carcinogenic potential. Over 2 decades, German toxicologist Elmar Richter, faculty member of the renowned Ludwig-Maximilians-University, Munich, received substantial financial support from the tobacco industry. Numerous publications show that his research findings supported the aims of the tobacco industry. In his commissioned work, he suggested that TSNA burden can be explained by misclassification of smokers or assay background levels caused by TSNA-like molecules from food. Other publications cast doubt on the relevance of animal testing for TSNAs to humans claiming a detoxifying effect of nicotine on the metabolism of TSNAs or suggesting that adducts of TSNAs are unsuitable as biomarkers of exposure to tobacco smoke. Conclusions: Economic interests of the tobacco industry have strongly influenced the research activity of Richter and his group. The publications of his working group about carcinogenic effects of TSNAs published between 1992 and 2009 should therefore not be regarded as independent. Scientists and policy makers should consider the long-standing and intensive inter-relation between certain toxicologists and the tobacco industry when assessing the research results and consider ignoring them.
Article
Besides 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL), [4-(methylnitrosamino)-1-(3-pyridyl)but-1-yl]-β-O-d-glucosiduronic acid (NNAL-Glu) is another important metabolite of the tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) which has been detected in the urine of tobacco users and non-smokers heavily exposed to sidestream cigarette smoke. In order to evaluate the toxicological significance of NNAL-Glu formation and excretion, the metabolism of [5-3H]-NNAL-Glu was studied in rats. Five male F344 rats were administered 3.7 mg/kg [5-3H]-NNAL-Glu by i.v. injection and the metabolites in urine analysed by HPLC. More than 90% of the radioactivity was excreted in urine within the first 24 h. Unchanged NNAL-Glu accounted for 81.2±3.1% of the total radioactivity; the remaining part of the dose appears to be deconjugated resulting in the urinary excretion of NNAL (3.6±1.7%) and its α-hydroxylation (11.5±2.2%) and N-oxidation (3.6±1.6%) products. The presence of α-hydroxylation products of NNAL-Glu in urine suggests that this NNK metabolite may be activated in vivo to carcinogenic intermediates.
Article
Full-text available
The bioactivation of the tobacco-specific nitrosamine, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), by microsomes from target organs was studied with an in vitro microsome-mediated DNA alkylation system. Mouse lung, rat lung, and rat nasal microsomes catalyzed a time- and protein-dependent DNA methylation by [methyl-3H]NNK with activities of 4.11, 0.95, and 137.4 pmol/mg DNA/mg protein/h, respectively. The DNA methylation of NNK catalyzed by all three microsomal systems was inhibited by cytochrome P-450 inhibitors, such as carbon monoxide and metyrapone, but not by the cyclooxygenase inhibitor, aspirin, or by prolonged preincubation in the absence of NADPH. The possible involvement of specific P450 isozymes was assessed by specific inhibitory antibodies. An anti-P450IIB1&2 antibody significantly inhibited the DNA methylation by 45 and 32% in mouse lung and rat lung, respectively, whereas anti-P450IA1 and anti-P450IIE1 antibodies failed to show significant inhibition. All antibodies showed no inhibition in rat nasal microsomes. Glutathione inhibited the DNA methylation in a concentration-dependent manner in all three microsomal systems. Phenethyl isothiocyanate (PEITC), at doses of 0.25 and 1.00 mmol/kg body weight, was given intragastrically 2 h before sacrifice to mice and 24 h before sacrifice to rats, respectively; both mouse and rat lung microsomal activities were inhibited by about 40 and 90% by the low- and high-dose PEITC treatments, respectively. The rat nasal microsomes were only inhibited by the high-dose PEITC treatment by about 40%. PEITC, 4-phenylbutyl isothiocyanate, and 6-phenylhexyl isothiocyanate all inhibited the microsome-mediated DNA methylation of NNK in vitro, with 4-phenylbutyl isothiocyanate and 6-phenylhexyl isothiocyanate being more potent than PEITC and the mouse lung microsomes more sensitive than the rat lung and nasal microsomes. All three microsomal systems were shown to catalyze the in vitro DNA pyridyloxobutylation by [5-3H]NNK. On an equal protein basis, the rat nasal microsomes were much more active in catalyzing the DNA pyridyloxobutylation.
Article
A gas chromatography, negative ion chemical ionization mass spectrometry (GC-NICI-MS) based assay for tobacco-specific nitrosamine adducts of DNA is described. The assay is based on the observation that acid hydrolysis of DNA from animals treated with tobacco-specific nitrosamines releases 4-hydroxy-1-(3-pyridyl)-1-butanone (HPB). HPB and the internal standard [4,4-D2]HPB are derivatized with pentafluorobenzoyl chloride and the resulting HPB-pentafluorobenzoate is purified by high-performance liquid chromatography prior to GC-NICI-MS analysis. DNA from human peripheral lung and tracheobronchial tissue, collected at autopsy, was analyzed for acid-released HPB. The mean HPB level (fmol/mg of DNA) for peripheral lung DNA was 11 +/- 16 (SD, n = 9) for smokers and 0.9 +/- 2.3 (n = 8) for nonsmokers. Mean adduct levels in tracheobronchus were 16 +/- 18 (n = 4) for smokers and 0.9 +/- 1.7 (n = 4) for nonsmokers. These are the first measurements of tobacco-specific nitrosamine-DNA adducts in humans. Further studies comparing the levels of DNA and globin adducts will provide a better understanding of the metabolic activation of tobacco-specific nitrosamines in humans and may provide a more accurate indication of an individual's risk of developing tobacco-related cancer.
Article
The tobacco-specific nitrosamine, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) induces tumor formation in the liver, lung, nasal cavity, and pancreas of rats. Metabolic activation is required for the tumorigenicity of this compound. The involvement of cytochrome P450 enzymes in NNK bioactivation was investigated in rats by studies with chemical inducers and antibodies against P450s. Liver microsomal enzymes catalyzed the formation of 4-oxo-1-(3-pyridyl)-1-butanone (keto aldehyde), 4-hydroxy-1-(3-pyridyl)-1-butanone (keto alcohol), 4-(methylnitrosamino)-1-(3-pyridyl-N-oxide)-1-butanone (NNK-N-oxide), and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL) from NNK. When the activity was expressed on a per nanomole P450 basis, treatments of rats with 3-methylcholanthrene (MC), phenobarbital (PB), pregnenolone 16-α-carbonitrile (PCN), Aroclor 1254 (AR), safrole (SA), and isosafrole (ISA) increased the keto aldehyde formation in liver microsomes 2.0-, 2.4-, 3.8-, 2.5-, 2.1-, and 1.8-fold, respectively; PB, AR, SA, and ISA increased the keto alcohol formation 1.7-, 1.3-, 2.0-, and 1.3-fold, respectively. The extents of induction were more pronounced when expressed on a per milligram protein basis, due to the higher microsomal P450 contents in the induced microsomes. The formation of NNK-N-oxide was markedly increased by PB and PCN and slightly increased by AR, SA, and ISA. However, the formation of NNAL, the major metabolite due to carbonyl reduction, was not increased by the treatments but was decreased by AR, ISA, and acetone (AC). The kinetic parameters of NNK metabolism by control, MC-, PB-, and PCN-induced liver microsomes were obtained. A panel of monoclonal (anti-1A1, -2B1, -2C11, and -2E1) and polyclonal (anti-1A2, -2A1, and -3A) antibodies were used to assess the involvement of constitutive hepatic P450 enzymes in NNK metabolism. Keto aldehyde formation was inhibited by anti-1A2 and anti-3A (about 15%) but not by others; the formation of keto alcohol was inhibited by anti-1A2, anti-2A1, and anti-3A (by 13–26%). In incubations with lung microsomes, the formation of keto aldehyde, keto alcohol, NNK-N-oxide, and NNAL were observed. With nasal mucosa microsomes, however, only keto aldehyde and keto alcohol formation were appreciable. SA and AC significantly decreased NNK metabolism in lung and nasal mucosa microsomes. Furthermore, SA inhibited the NNK oxidative metabolism in lung and nasal mucosa microsomes in vitro. AC significantly inhibited the NNK oxidative metabolism in lung microsomes in vitro, but not in nasal mucosa microsomes. The present results demonstrate the important role of P450 enzymes in the metabolism of NNK. Several P450 isozymes can catalyze the oxidation of NNK with different but overlapping regioselectivities and P450 inducers affect the metabolism of NNK differently in the liver, lung, and nasal mucosa.
Article
Three male marmoset monkeys ( Callithrix jacchus ) were injected i.v. with the tobacco-specific carcinogen [2'-14C]N'-nitrosonornicotine (NNN) (20.3 μmol&sol;kg body weight) or [carbonyl-14C]4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) (18.8 or 420 μmol&sol;kg body weight). They were sacrificed 4 h later. Tissue distribution was studied in two monkeys by whole-body autoradiography and by computer-assisted densitometric analysis of the autoradiograms. The autoradiograms showed a high level of radioactivity in the liver, nasal mucosa, kidneys, melanin of the eyes, hair-follicles of the skin and in the ceruminous ear glands of the monkeys. Total level of radioactivity was 5.7 times higher in the liver of the [carbonyl-14C]NNK-injected monkey than in that of [2'-14C]NNN-injected monkey. Washing the sections with trichloroacetic acid and organic solvents selectively removed free metabolites leaving metabolites bound to cellular macromolecules. Level of bound metabolites was 1.5 times higher in the nasal mucosa than in the liver of the [2'-14C]NNN monkey. Levels of bound metabolites were similar in the liver of NNN-and NNK-treated monkeys. The results indicate that the liver and nasal mucosa of C. jacchus can activate NNN and NNK to alkylating species. Unbound metabolites present in the liver, lung, kidneys, eye, blood and urine were extracted and separated by h.p.l.c. Hydroxylation of the carbons α to the N-nitroso group of NNN were the major metabolic pathways. Unmetabolized NNN was the major radioactive component in the liver, lung, eye and blood. Reduction of the carbonyl of NNK yields 4-(methylnitrosamino)-1-(3-pyridyl)butan-1-ol (NNAI). NNAI was present in all tissues analyzed and was the major radioactive component in the eye and stomach lumen. It was also excreted in the urine. NNK and NNAI were metabolized by α-carbon hydroxylation. These results suggest that in C. jacchus , NNN, NNK and NNAI are activated to alkylating species by α-carbon hydroxylation. In the third monkey injected with NNK, DNA methylation was observed in the liver and nasal mucosa but not in the lung and kidneys. Pulmonary tissues of C. jacchus , unlike those of F344 rats, do not have the enzymic capacities to activate NNK to methylating species.
Article
4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), a potent tobacco-specific carcinogen in animals, has been linked to tobacco-related cancers in humans. The cytochrome(s) P-450 (P-450) responsible for the metabolic activation of NNK in humans has not been identified. The present work investigated the ability of human lung and liver microsomes and 12 forms of human P-450, expressed in Hep G2 (hepatoma) cells, to metabolize NNK. Of the 12 P-450 forms, P-450 1A2 had the highest activity in catalyzing the conversion of NNK to the keto alcohol, 4-hydroxy-1-(3-pyridyl)-1-butanone. P-450s 2A6, 2B7, 2E1, 2F1, and 3A5 also had measurable activities in the formation of keto alcohol. The apparent Km and Vmax for the formation of keto alcohol in the P-450 1A2-expressed Hep G2 cell lysate were 309 microM and 55 pmol/min/mg protein, respectively. 4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanol, a reductive product, was the major metabolite formed, whereas the formation of keto alcohol and its aldehyde and acid derivatives (all alpha-hydroxylation products) constituted approximately 1% of the initial amount of NNK in P450-expressed Hep G2 cell lysate. A similar metabolite pattern was observed with human lung or liver microsomes. In human lung microsomes, the apparent Kms for the formation of 4-hydroxy-4-(3-pyridyl)butyric acid, 4-oxo-1-(3-pyridyl)-1-butanone, NNK-N-oxide, and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol were 526, 653, 531, and 573 microM, respectively; the formation of keto alcohol was not observed. For human lung microsomes, there was no sex-related difference in NNK metabolism. Carbon monoxide (90% atmosphere) significantly inhibited the metabolism of NNK in human lung and liver microsomes. 7,8-Benzoflavone, an inhibitor of P-450s 1A1 and 1A2, had no effect on NNK metabolism in human lung microsomes but decreased the formation of keto alcohol by 47% in human liver microsomes. Similarly, antibodies against human P-450s 1A2 and 2E1 decreased keto alcohol formation by 42% and 53%, respectively, in human liver microsomes but did not affect NNK metabolism in lung microsomes. Inhibitory antibodies against P-450s 2A1, 2C8, 2D1, or 3A4 had little or no effect on the metabolism of NNK in human liver or lung microsomes.(ABSTRACT TRUNCATED AT 400 WORDS)
Article
Rabbit nasal olfactory and respiratory microsomes were found to catalyze the alpha-hydroxylation of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) with specific activities of 262 and 136 pmol/min/mg protein in the formation of keto aldehyde, and of 318 and 190 pmol/min/mg protein in the formation of keto alcohol respectively. The formation of NNK-N-oxide was observed in experiments with rabbit olfactory and respiratory microsomes, but not with rat nasal microsomes. However, the rat nasal microsomes had higher activity in catalyzing the alpha-hydroxylation of NNK. In a reconstituted system, rabbit P450NMa, a major constitutive P450 isozyme in nasal microsomes, displayed high activities in the formation of the keto aldehyde and the keto alcohol with apparent Km values of 15 and 9 microM respectively. In comparison, rabbit olfactory specific P450NMb had a low activity in catalyzing the formation of keto aldehyde (Km = 186 microM) and no activity in the formation of keto alcohol. The P450NMa-catalyzed oxidation of NNK was inhibited by nicotine and diallyl sulfide. Kinetic studies indicated that nicotine is a competitive inhibitor. These results demonstrate that enzymes in rabbit nasal microsomes, especially P450NMa, efficiently catalyze the bioactivation of NNK.
Article
The tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) is a potent pancreas carcinogen in rats. The biliary excretion of NNK was therefore studied in anesthetized female Sprague-Dawley rats following i.p. administration of 0.7 mumol/kg [carbonyl-14C]NNK. The concentration of radioactivity peaked within 30 min and decreased thereafter exponentially. Cumulative excretion of radioactivity reached a plateau at 6-9% of the total dose. HPLC analysis revealed the presence of 4-hydroxy-4-(3-pyridyl)butyric acid (hydroxy acid), 4-oxo-4-(3-pyridyl)-butyric acid (keto acid), 4-(methylnitrosamino)-1-(3-pyridyl)-1-butyl beta-D-glucopyranosiduronic acid (NNAL Glu), 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL) and NNK. NNAL Glu was the major metabolite contributing 34 +/- 4% of total radioactivity in bile at 30 min and 58 +/- 4% at 5 h. The percentage of acidic metabolites remained constant at approximately 20%. In contrast, the percentage of NNK and NNAL decreased within the first 2 h to < 5% and < 10% respectively. The elimination kinetics of NNK and its metabolites fitted into a one-compartment model with a half-life of 37 min for NNK, 52 min for NNAL and 110 min for NNAL Glu and acidic metabolites. In three rats dosed with 240 mumol/kg NNK i.p., the concentration of radioactivity peaked after 1-2 h and decreased very slowly thereafter. After 5-8 h a total of 12-17% of the dose has been excreted in the bile with no indication of a plateau. At all time points NNAL Glu was the major metabolite contributing up to 95% of total radioactivity in bile. The percentage of acidic metabolites was < 5% throughout the experiment. Whereas NNK contributed one-third of the radioactivity at 30 min and decreased rapidly, the percentage of NNAL in bile remained rather constant at approximately 5-10%. In conclusion, the detection of NNK, NNAL and NNAL Glu gives support to the hypothesis that tobacco-specific carcinogens could reach the pancreas retrograde from the bile, especially at high NNK concentrations.
Article
An acetyltransferase-overexpressing strain of Salmonella typhimurium (NM2009) has been used to investigate roles of human liver microsomal cytochrome P450 (P450) enzymes in the activation of carcinogenic nitrosamine derivatives, including N-nitrosodialkylamines and tobacco-smoke-related nitrosamines, to genotoxic products. Studies employing correlation of activities with several P450-dependent monooxygenase reactions in different human liver samples, inhibition of microsomal activities by antibodies raised against human P450 enzymes and by specific P450 inhibitors, and reconstitution of activities with purified P450 enzymes suggest that the tobacco-smoke-related nitrosamines 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL) and N-nitrosonornicotine (NNN) as well as N-nitrosodimethylamine (NDMA) and N-nitrosodiethylamine (NDEA) are oxidized to genotoxic products by different P450 enzymes, particularly P450 2E1 and 2A6. The activation of NDMA and NNN by liver microsomes was suggested to be catalyzed more actively by P450 2E1 than by other P450 enzymes because the activities were well correlated with NDMA N-demethylation and aniline p-hydroxylation in different human samples, and purified P450 2E1 had the highest activities in reconstituted monooxygenase systems. The relatively high contribution of P450 2A6 to the activation of NDEA and NNK was supported by the correlation seen with coumarin 7-hydroxylation in human liver microsomes, and antibodies raised against P450 2A6 inhibited both activities by approximately 50%. P450 3A4, 2D6 and 2C enzymes appear not to be extensively involved in the activation of these nitrosamines as judged by several criteria examined. Thus, this work indicates that several P450 enzymes, particularly P450 2E1 and 2A6, catalyze metabolic activation of nitrosamine derivatives including N-nitrosodialkylamines and tobacco-smoke-related nitrosamines in human liver microsomes.
Article
Tobacco-specific nitrosamines are derived from nicotine and related tobacco alkaloids and can be detected in tobacco products as well as in mainstream and sidestream smoke. Two of them, N-nitrosonornicotine and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone, are strong carcinogens in laboratory animals. Because of its organospecificity for the lung, the latter is considered to be a causative factor in tobacco-related human lung cancer. Upon metabolic activation both nitrosamines give rise to a common reactive intermediate binding to macromolecules such as DNA and haemoglobin and hydrolysing to 4-hydroxy-1-(3-pyridyl)-1-butanone. Because of easy access to large quantities of haemoglobin from blood samples, it is most suitable for biomonitoring human exposure to tobacco-specific nitrosamines. A highly sensitive analytical method for determination of femtogram amounts of 4-hydroxy-1-(3-pyridyl)-1-butanone provides an approach to assess individual exposure to active and passive smoking.
Article
The relative importance of the two alpha-hydroxylation pathways in the tumorigenicity of the tobacco-specific nitrosamine, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), was examined in the A/J mouse lung. Methyl hydroxylation, which results in DNA pyridyloxobutylation, was investigated with 4-(acetoxymethylnitrosamino)-1-(3-pyridyl)-1-butanone (NNKOAc) and N'-nitrosonornicotine. Methylene hydroxylation, which leads to DNA methylation, was studied by using acetoxymethyl-methylnitrosamine (AMMN). The tumorigenic activities of these compounds were compared to that of 10 mumol NNK at doses that yielded similar or greater adduct levels 24 h after exposure. The methylating agent AMMN was more tumorigenic than the pyridyloxobutylating agents, NNKOAc and N'-nitrosonornicotine. NNKOAc enhanced the tumorigenic activity of AMMN when the two compounds were given in combination. These results suggested that DNA methylation was more important than DNA pyridyloxobutylation in A/J mouse lung tumor induction by NNK and that pyridyloxobutylation enhanced the activity of the methylation pathway. However, the tumorigenicity of 10 mumol NNK could not be reproduced by AMMN +/- NNKOAc at doses that yielded similar levels of DNA adducts 24 h after exposure. Therefore, a second study was conducted in which the persistence of O6-methylguanine in lung DNA following various doses of NNK or AMMN +/- NNKOAc was compared to the tumorigenicity of these treatments. A strong correlation was observed between lung tumor yield and levels of O6-methylguanine at 96 h for NNK and AMMN +/- NNKOAc (r = 0.98). The ability of NNKOAc to increase the tumorigenic activity of AMMN was attributed to its ability to enhance the persistence of O6-methylguanine in lung DNA. These results demonstrate that the formation and persistence of O6-methylguanine are critical events in the initiation of A/J mouse lung tumors by NNK. They also suggest that DNA pyridyloxobutylation by NNK can increase the persistence of this promutagenic base in lung DNA.