• Nie Znaleziono Wyników

Combined antioxidant (beta-carotene, alpha-tocopherol and ascorbic acid) supplementation increases the levels of lung retinoic acid and inhibits the activation of mitogen-activated protein kinase in the ferret lung cancer model.

N/A
N/A
Protected

Academic year: 2021

Share "Combined antioxidant (beta-carotene, alpha-tocopherol and ascorbic acid) supplementation increases the levels of lung retinoic acid and inhibits the activation of mitogen-activated protein kinase in the ferret lung cancer model."

Copied!
10
0
0

Pełen tekst

(1)

Combined antioxidant (b-carotene, a-tocopherol and ascorbic acid) supplementation

increases the levels of lung retinoic acid and inhibits the activation of

mitogen-activated protein kinase in the ferret lung cancer model

Yuri Kim

1

, Nalinee Chongviriyaphan

1,2

, Chun Liu

1

,

Robert M.Russell

1

and Xiang-Dong Wang

1,

1Nutrition and Cancer Biology Laboratory, Jean Mayer United States Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA

2Present address: Division of Nutrition, Department of Pediatrics, Faculty of Medicine at Ramathibodi Hospital, Mahidol

University, Rama 6 Road, Bangkok, Thailand 10400

To whom correspondence should be addressed at: The Nutrition and

Cancer Biology Laboratory, Jean Mayer United States Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA.

Tel:+1 617-556-3130; Fax: +1 617-556-3344;

Email: xiang-dong.wang@tufts.edu

Interactions among

b-carotene (BC), a-tocopherol (AT)

and ascorbic acid (AA) led to the hypothesis that using

a combination of these antioxidants could be more

beneficial than using a single antioxidant alone,

particu-larly against smoke-related lung cancer. In this

investiga-tion, we have conducted an animal study to determine

whether combined BC, AT and AA supplementation

(AOX)

protects

against

4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced lung carcinogenesis

in smoke-exposed (SM) ferrets. Ferrets were treated

for 6 months in the following four groups: (i) control,

(ii) SM

+ NNK, (iii) AOX and (iv) SM + NNK + AOX.

Results showed that the combined AOX supplementation

(i) prevented the SM

+ NNK-decreased lung

concentra-tions of retinoic acid (RA) and BC; (ii) inhibited the

SM

+ NNK-induced phosphorylation of Jun N-terminal

kinase

(JNK),

extracellular-signal-regulated

protein

kinase (ERK) and proliferating cellular nuclear antigen

proteins in the lungs of ferrets; and (iii) blocked the SM

+ NNK-induced up-regulation of total p53 and Bax

proteins, as well as phosphorylated p53 in the lungs of

ferrets. In addition, there were no lesions observed in

the lung tissue of ferrets in the control and/or the AOX

groups after 6 months of intervention, but combined

AOX supplementation resulted in a trend toward lower

incidence of both preneoplastic lung lesions and lung

tumor formation in SM

+ NNK + AOX group of ferrets,

as compared with the SM

+ NNK group alone. These

data indicate that combined AOX supplementation could

be a

useful

chemopreventive strategy

against lung

carcinogenesis through maintaining normal tissue levels

of RA and inhibiting the activation of mitogen-activated

protein

kinase

pathways,

cell

proliferation

and

phosphorylation of p53.

Introduction

Lung cancer has been the leading cause of cancer death in both

men and women in the United States for the past 10 years.

Cigarette smoking is the main risk factor for the development

of lung cancer and avoidance of tobacco products is the best

way to prevent tobacco-related cancers. However, the

addict-ive power of nicotine is strong and exposure to environmental

tobacco smoke (e.g. second-hand smoke) persists. Protection

by consuming a healthy diet may be an effective way to protect

against the harmful effects of tobacco smoke and reduce

lung cancer risk. Observational epidemiologic studies have

consistently demonstrated that individuals eating more fruits

and vegetables, which are rich in carotenoids and people

having higher serum

b-carotene (BC) levels have a lower

risk of cancer, particularly lung cancer (1). In contrast, two

intervention trials, the Alpha-Tocopherol, Beta-carotene

Can-cer Prevention Trial (ATBC) in Finland (2) and the

b-Carotene

and Retinol Efficacy Trial in the United States (3)

demon-strated an increased risk of lung cancer in heavy smokers

and asbestos workers if they consumed supplements containing

BC. A possible explanation of why high doses of BC in

smokers caused more lung cancer is that BC is susceptible

to oxidation in the free radical rich, antioxidant poor

environ-ment of the lungs of cigarette smokers (4,5). We found that

cigarette smoke enhanced the formation of oxidative excentric

cleavage products of BC in ferrets (6), which facilitate the

binding of benzo[a]pyrene metabolites to DNA (7) and induce

several cytochrome P450 (CYP) enzymes (such as CYP1A1/2

and CYP2A1, which activate tobacco-smoke procarcinogens)

in the lungs of ferrets (8). Furthermore, we have shown that

the induction of CYP enzymes in the lungs of ferrets exposed

to cigarette smoke and/or high dose of BC (30 mg/day)

enhances retinoic acid (RA) catabolism, which provides an

additional explanation for enhanced lung carcinogenesis (4,8).

Previous in vitro studies suggest that there are strong in vitro

interactions among BC,

a-tocopherol (AT) and ascorbic acid

(AA) in terms of mutual beneficial protection against oxidative

damage (9–11). AT and AA have the capability to regenerate

BC from its radical cation (9), thus preventing BC from being

further oxidized. In addition, BC can enhance AT antioxidant

efficiency by regenerating AT from its radical cation (10).

We have shown that the in vitro addition of both AT and

AA into the incubation mixture of BC with smoke-exposed

ferret lung tissue can inhibit the production of excentric

cleavage metabolites of BC, but increase the levels of retinal

and RA (12). Although pre-treatment of human lung cells with

both vitamin E and BC has been shown to provide protection

against DNA strand breaks induced by tobacco-specific

nitrosamines (11), the combination of BC (20 mg/day) and

vitamin E (50 mg/day) were not found to be protective against

smoke-related lung cancer in the ATBC study. However,

vitamin C, which facilitates both recycling and stability of

AT and BC as well as converting BC into RA (12–14), was

Abbreviations:AA, ascorbic acid; AT,a-tocopherol; BC, b-carotene; ERK,

extracellular-signal-regulated protein kinase; JNK, Jun N-terminal kinase; MAPK, mitogen-activated protein kinase; PCNA, proliferating cellular nuclear antigen; RA, retinoic acid.

(2)

not used in the ATBC study and would be expected to be low

in this population of heavy smokers. In a follow-up study

from the first National Health and Nutrition Examination

Survey, the subjects in the highest quartiles of carotenoid,

vitamin E and vitamin C consumption had a 68% lower risk

of lung cancer than those in the lowest quartiles of all three

nutrients (15). A recent prospective cohort study exploring

the question of whether combinations of dietary antioxidants

affect lung cancer morbidity and mortality in male smokers has

shown that men with the highest antioxidant index scores had

a significantly lower (16%) risk of lung cancer than men with

the lowest scores (16). Although it is possible that using a

combination of these antioxidants would be advantageous in

antioxidant protection against smoke-related lung cancer, more

evidence supporting the hypothesis and exploration of the

possible mechanism(s) involved are needed.

Jun N-terminal kinase (JNK) and

extracellular-signal-regulated protein kinase (ERK) belong to mitogen-activated

protein kinase (MAPK) family and are activated by

phos-phorylation in response to many extracellular stimuli and

environmental stress factors (e.g. smoke exposure) and may

play an important role in carcinogenesis (17–19). JNK

was shown to phosphorylate c-Jun on sites serine-63 and

serine-73 (20) and increase AP-1 transcription activity (21)

and, eventually, mediate cell proliferation and apoptosis.

ERK induced c-Jun through phosphorylation and activation

of AP-1 component ATF1 at serine-63 (22). In addition,

JNK and ERK mediate phosphorylation of the p53 tumor

suppressor (19,23). Phosphorylation of p53 plays a role in

cell accumulation in G

1

phase of the cell cycle and signals

oxidative stress in cells. Phospho-p53 appears to be expressed

in cells undergoing apoptosis. Serine-15 in human p53 is

sensitive to oxidative stress-induced phosphorylation (24).

Previously, we have shown that high dose of BC

supplementa-tion promotes the smoke-induced phosphorylasupplementa-tion of JNK

and p53 (25). However, it is not known whether BC combined

with AT and AA supplementation can affect the MAPK

signaling pathway or phosphorylation of p53 and its

downstream gene Bax.

The ferret (Mustela putorius furo) has been shown to be

an excellent model for studying carotenoid absorption

and metabolism (6,26–30). Recently, we have performed a

6-month in vivo study in ferrets exposed to both tobacco

smoke and a carcinogen

[4-(N-methyl-N-nitrosamino)-1-(3-pyridyl)-1-butanone, NNK] found in cigarette smoke (31).

Based on the close similarity of the animal’s lung tumor

pathology to humans, we have demonstrated the ferret to be

a good model for lung carcinogenesis. In the present report,

we conducted a ferret study to determine whether combined

BC, AT and AA supplementation protects against altered

BC metabolism, activation of the MAP kinase pathway and

chemical carcinogen-induced carcinogenesis in the lungs

of smoke-exposed ferrets.

Materials and methods

Animals, diet and study group

Adult male ferrets (1.2–1.4 kg) from Marshall Farms (North Rose, NY) were housed in an American Association of Accreditation of Laboratory Animal Care-accredited animal facility at the Human Nutrition Research Center on Aging at Tufts University (HNRCA), fed a semipurified ferret diet (Research Diets, New Brunswick, NJ) and provided water ad libitum. Accord-ing to our previous study, the average daily food intake of the ferret, with an average body weight of 1.3 kg, is 80 g/day. In 80 g of this diet, a ferret gets

a negligible amount of BC, 0.43 mg of retinyl palmitate, 2 mg AT and no AA. All animals were quarantined for a minimum of 1 week to ascertain their health status before the experiment began. During the experimental period, ferret body weights were recorded weekly. After the experimental period, all ferrets were terminally exsanguinated under deep isoflurane anesthesia. Tissues were collected and stored at80C until analyzed. All experimental procedures carried out on the ferrets were reviewed and approved by the Animal Care and Use Committee of the HNRCA.

Short-term (6-week) study. Thirty-six ferrets were randomly assigned to four groups for a 6-week experiment as follows: (i) control (neither vitamin

supplemented nor exposed to cigarette smoke), n¼ 9; (ii) smoke exposed

(SM) without any supplementation, n¼ 9; (iii) smoke exposed plus low dose BC (LBC, 0.48 mg/kg body wt/day), AT (22 mg/kg body wt/day) and AA (3 mg/kg body wt/day) supplementation, n¼ 9; and (iv) smoke exposed plus high dose BC (HBC, 2.4 mg/kg body wt/day), AT (22 mg/kg body wt/day) and

AA (3 mg/kg body wt/day) supplementation, n¼ 9.

Long-term (6-month) study. Forty-four male ferrets were randomly assigned to four groups as follows: (i) control (neither vitamin supplemented

nor exposed to cigarette smoke), n ¼ 9; (ii) smoke-exposed plus

NNK-treated (SM + NNK), n ¼ 12; (iii) combined antioxidant supplemented

[AOX, BC (0.85 mg/kg body wt/day), AT (22 mg/kg body wt/day) and AA

(3 mg/kg body wt/day)], n¼ 9; and (iv) SM + NNK treated plus combined

AOX supplemented, n¼ 14.

Cigarette smoke exposure and NNK treatment

We conducted a short-term (6-week) study using the ferret model exposed to smoke only and a long-term (6-month) study using the ferret model exposed to both smoke and NNK treatment, as described previously (31). In brief, we exposed ferrets to smoke from non-filtered cigarettes (Standard Research Cigarettes, Type 2R4F, Tobacco-Health Research Institute, University of Kentucky, Lexington, KY) (6). Ferrets were exposed to cigarette smoke twice (30 min each time) per day throughout the experimental period. We showed that this amount of smoke exposure in the ferret is similar to that found in humans smoking one pack of cigarettes per day. For the carcinogen treatment, 50 mg NNK/kg body weight was given by i.p. injection at 4-week intervals for a total of 4 doses over 4 months. This dose was based on the report that mink receiving a single of dose (150 mg) of tobacco-specific N-nitrosamine, N0-nitrosonornicotine (NNN) did not show any toxic effects (32). Control groups were given a sham injection of normal saline. The sham-exposed ferrets were housed in a separate room and went through the exact same procedures as the smoke-exposed animals, except that they received neither smoke nor carcinogen exposure.

BC, AT and AA supplementation

Natural all-trans BC (Cognis, Cincinnati, OH) and all-rac-a-tocopherol acetate (Roche Vitamins, NJ) were dissolved in 1 ml of corn oil and fed orally (not gavage) to the supplemented ferrets every morning. Ferrets like to eat corn oil and lick it spontaneously. AA (Roche Vitamins) was freshly prepared by dissolving it in distilled water and then fed orally to the supplemented ferrets every morning at a dose of 3 mg AA/kg body wt/day, which is equivalent to 210 mg AA/day in a 70 kg man. Ferrets in the control group

and SM+ NNK group were fed the basal diet plus 1 ml corn oil without

antioxidants. In the short-term study, we used BC at low (0.48 mg BC/kg body wt/day) and high (2.4 mg BC/kg body wt/day) doses, which had been previously shown to be protective and harmful, respectively, in smoke-exposed ferrets (33). Based on our previous study (26), the total absorption of intact BC by ferrets is5 times lower than that in the human. The LBC at 0.48 mg BC/kg body wt/day in the ferret is therefore equivalent to 6 mg BC/day in a 70 kg man. The HBC at 2.4 mg BC/kg body wt/day in the ferret is equivalent to 30 mg BC/day in a 70 kg man. Based on the result from the short-term study, 0.85 mg BC/kg body wt/day in ferrets (equivalent to 12 mg BC/day in a 70 kg man) was chosen for the long-term study in order to maintain the lung RA concentrations at a normal level. From these short-term data, the final sample size of ferrets chosen in the long-term study was one that would allow a 80% chance of detecting statistically significant differences among the groups at a 0.05 level of significance, assuming a 50% reduction of both the RA level and the incidence of lung squamous metaplasia in the

lungs of the SM+ BC + AT + AA group as compared with the SM exposed

alone group.

Tissue extraction and HPLC analysis

BC (33), AT (30,34), AA (35) and retinoids including retinyl palmitate, retinol and RA in lung tissue homogenates were measured by HPLC and UV detection, as described previously with some modifications. A total of 0.2 g of lung tissue (wet weight) in a mixture of normal saline and methanol (2:1, v/v) was homogenized on ice for 30 s, followed by addition of 100ml

(3)

of internal standard (retinyl acetate). After adding 5 ml of chloroform:methanol (2:1, v/v), the mixture was vortexed and centrifuged for 10 min at 800 g at 4C. Four milliliter of hexane was added after the lower layer had been collected.

The chloroform and hexane layers were evaporated under N2 and a 50ml

aliquot of the extract reconstituted with ethanol was injected into a gradient reverse-phase HPLC system, consisting of a Waters 2695 separation module, a Waters 2996 photodiode array detector (Waters Corporate, Milford, MA)

and a 0.46 · 8.3 cm Pecosphere-3 C18 cartridge column (Perkin-Elmer

Analytical Instruments, Shelton, CT). The HPLC mobile phase consisted of acetonitrile:tetrahydrofuran:water (50:20:30, v/v/v, 1% ammonium acetate in water) as solvent A and acetonitrile:tetrahydrofuran:water (50:44:6, v/v/v, 1% ammonium acetate in water) as solvent B at flow rate of 1 ml/min. Individual compounds were identified by coelution with standards and were quantified by determining peak areas in the HPLC chromatograms calibrated against known amount of standards. Vitamin levels were corrected for extraction loss based on the recovery of the internal standard. All procedures were carried out under red light.

Histopathology and immunohistochemistry

The histopathology examination and procedures used for the lung have been described elsewhere (31,33). The WHO International Association for the Study of Lung Cancer classification of lung and pleural tumors (36,37) and the WHO Histological Classification of Tumors of the Respiratory System for domestic animals (37) were used as guidelines for histopathological diagnoses. The right upper lobe of each ferret lung was inflated and fixed by an intratracheal instillation of 10% formalin. The samples were then embed-ded in paraffin. The lung sections, 5mm in thickness, were made using an AO microtome and stained with hematoxylin and eosin. Lung lesions were independently examined and diagnosed by a pathologist and by two invest-igators who were blinded to the treatment groups. If any premalignant or malignant lesions were observed (histological examination plus confirmation by immunohistochemistry) in a ferret, the ferret was considered as ‘positive’. Otherwise, the animal was considered ‘negative’. Localization of phospho-p53 (serine-15, Cell signaling, Beverly, MA) in a lung sections from a tumor

bearing animals in the SM+ NNK group was analyzed by using

immunohis-tochemistry, as described previously (33). Western blot analysis

Western blot analysis of protein levels was carried out for JNK, ERK, Bax, phospho-JNK and phospho-ERK. Briefly, lung tissues were incubated in

extraction buffer (25 mM HEPES, 300 mM NaCl, 1.5 MgCl2, 0.2 mM

EDTA, 0.05% Triton X-100, and 20 mMb-glycerophosphate and a mixture

of protease inhibitors) with agitation at 4C for 30 min. The mixture was then centrifuged and supernatants were collected. Western blot analysis for p53, phospho-p53 at serine-15 and proliferating cellular nuclear antigen (PCNA) were carried out using nuclear protein extracts from the lungs of ferrets. The lung tissues were homogenized gently with ice-cold buffer A [10 mM Tris–HCl (pH 7.5), 10% glycerol, 10 mM KCl, 10 mM monothioglycerol with a mixture of protease inhibitors] and nuclei were collected by centrifu-gation for 30 min at 3200 g. Nuclear pellets were solubilized in buffer B [10 mM Tris–HCl (pH 7.5), 10% glycerol, 600 mM KCl, 1 mM DTT, 10 mM monothioglycerol with a mixture of protease inhibitors]. The extracts were centrifuged for 30 min at 100 000 g; the resulting supernatants are referred to as the nuclear extract. SDS–PAGE and western blot analysis were carried out according to the standard protocols using antibodies against total JNK, phospho-JNK, PCNA (Santa Cruz Biotechnology, Santa Cruz, CA). Antibodies against phospho-p53 (serine-15), total ERK, phospho-ERK, Bax were purchased from Cell Signaling and antibody against p53 was purchased from Calbiochem (San Diego, CA).

Statistical analysis

All results are expressed as means ± SEM. Data were analyzed by ANOVA followed by Tukey’s honest significant difference test, unless otherwise indicated. Differences were considered significant at P< 0.05.

Results

Lung squamous metaplasia and lung concentration of RA in

ferrets after 6 weeks of treatment

There were no significant differences in ferret body weight

among the treatment groups at the beginning, during,

or after the 6-week experiment (data not shown). The

results of the pathological examination for the effect of

com-bined antioxidant supplementation on smoke-induced lung

squamous metaplasia in ferrets after 6 weeks of treatment

are presented in Table I. There was one animal in the control

group having lung squamous metaplasia, probably due to

spon-taneous occurrence. Although there were no significant

differ-ences in the incidence of lung squamous metaplasia among the

control, the SM alone and the SM

+ HBC + AT + AA groups,

the number of ferrets with squamous metaplasia in the SM

+

LBC

+ AT + AA group was significantly lower than that in the

SM group (Table I). Lung RA concentrations were

signific-antly lower in the SM group than in the control group, as we

reported before (33). Although LBC in the presence of AT and

AA did restore the lung RA concentration to that of the control

group, the lung RA concentration was significantly higher in

the SM

+ HBC + AT + AA group as compared with the control

group (Table I).

Concentration of antioxidants (BC, AT and AA) and retinoids

(RA, retinol and retinyl palmitate) in lung tissue of ferrets

after 6 months of treatment

The AOX supplemented groups showed significantly higher

lung BC after 6 months, as compared to non-supplemented

groups (Table II). Interestingly, in the presence of AT and

AA, the lung BC concentration in the SM

+ NNK + AOX

was maintained at the level equal to that of the AOX alone

group. AT levels in lung tissues of ferrets were significantly

higher in AOX group than in the control group (2.2-fold

difference) (Table II). The concentration of AT in lung tissues

was not affected by SM exposure plus NNK treatment in the

non-supplemented group, whereas AT concentration was

significantly higher in SM

+ NNK + AOX group, compared

with the AOX alone group. There were significantly lower

lung levels of both reduced AA (28% lower) and total AA

(38% lower) among smoke-exposed plus NNK-treated ferrets,

as compared with control animals with or without AOX

supplementation. However, the level of either reduced

or total AA lung concentrations was not affected by AOX

supplementation.

RA concentrations in lung tissue were significantly lower

(90%) in the SM

+ NNK group as compared with the control

group. However, the RA concentration in the lungs of ferrets

of the AOX group was higher (2 fold difference) than that of

the control group. The RA concentration was restored to

Table I. The effect of 6 weeks of smoke-exposure (SM) with or without

antioxidants (BC, AT and AA) supplementation on lung squamous metaplasia and lung concentration of RA in ferrets

Number of ferrets having lung squamous metaplasia/the total number in each group

Lung concentration of RA (pmol/g) Control 1/9 (11%)a,b 24.17 ± 3.89a SM 4/9 (44%)a 10.52 ± 0.83b SM+ LBC + AT + AA 0/9 (0%)b 14.55 ± 1.51b SM+ HBC + AT + AA 1/9 (11%)a,b 48.01 ± 13.34c

Number of ferrets with squamous metaplasia/the total number of ferrets in each group (%). SM, smoke-exposed; LBC, low dose (equivalent

daily intake of 6 mg BC in a 70 kg man) ofb-carotene supplemented;

HBC, high dose (equivalent daily intake of 30 mg BC/day in a 70 kg

man) ofb-carotene supplemented; AT, a-tocopherol dose (100 mg

AT/day) supplemented; AA, ascorbic acid (210 mg AA/day); RA, retinoic acid.

a,b,cFor a given column, data not sharing a common superscript letter are statistically significantly different at P< 0.05 (Fisher’s Exact test for the incidence of lung squamous metaplasia).

(4)

control levels in the SM

+ NNK groups by BC supplementation

in the presence of AT and AA (Table II). We observed

no significant differences in the concentrations of lung retinol

and/or retinyl palmitate among the four groups (Table II).

Phosphorylated JNK, phosphorylated ERK and PCNA protein

expression in the lung tissue of ferrets

We observed no difference in protein levels of total JNK

among four treatment groups (Figure 1A). As compared

with the control group, phospho-JNK protein levels in the

smoke-exposed plus NNK-treated group were higher (58%).

This up-regulation of JNK phosphorylation in SM

+ NNK

group was significantly inhibited by AOX treatment.

Further-more, ERK phosphorylation was significantly higher in SM

+

NNK treatment (2.2-fold difference), as compared with

the control or the AOX group (Figure 1B); whereas, AOX

supplementation prevented the SM+NNK-induced ERK

phos-phorylation. The levels of total ERK were not different among

four groups. Since MAPK phosphorylation mediates cell

proliferation, PCNA protein expression in lung tissue was

examined to evaluate a potentially higher cell proliferation

with SM

+ NNK treatment. Compared with the control

group or the AOX treated group, PCNA expression was

significantly higher (36%) in the SM

+ NNK group

(Figure 1C). This up-regulated PCNA expression in the

SM

+ NNK group was inhibited by AOX supplementation

(Figure 1C).

Levels of p53, phosphorylated p53 (serine-15) and Bax in the

lung tissue of ferrets

There was higher (50–63%) total p53 protein level in the

group exposed to SM

+ NNK (Figure 2A) compared with

the control group, the AOX group and the SM

+ NNK +

AOX group. No differences in total p53 protein levels were

observed among the control groups, the AOX group and the

SM

+ NNK + AOX group. Because p53 phosphorylation is

mediated by MAPKs such as JNK and ERK, we examined

p53 phosphorylation. As compared with the control and the

AOX groups, p53 protein phosphorylation at serine-15 was

higher in the groups exposed to SM

+ NNK (2-fold

differ-ence) (Figure 2B). This SM

+ NNK-induced phosphorylation

of p53 was blocked by AOX supplementation. No difference

in phopho-p53 protein levels were observed between the

AOX group, the SM

+ NNK + AOX group and the control

group. Further analysis of p53 phosphorylation using the

antibody against phosphorylated p53 showed significantly

higher

phospho-p53

expression

in

the

tumor

region

(Figure 3A and C), as compared with non-tumor regions

(Figure 3A and B). These data indicate that higher p53

phosphorylation in the tumor region may explain the higher

levels of p53 phosphorylation in SM

+ NNK group, as

com-pared with the control group. Since phosphorylation of

p53 stabilizes and activates p53, we examined Bax, a major

downstream gene of p53. Bax protein expression was

signi-ficantly higher (70%) in the SM

+ NNK group, compared

with the control group (Figure 2C). AOX supplementation

prevented this induction of Bax by SM

+ NNK treatment.

No difference in Bax protein expression levels were observed

between the SM

+ NNK + AOX group, the control group or

the AOX treated group.

Lung preneoplastic and neoplastic lesions

The treatment of ferrets with combined cigarette smoke and

NNK resulted in both the formation of preneoplastic lesions

and neoplastic lesions (Figure 4). The preneoplastic lesions

included both squamous dysplasia (Figure 4A) and atypical

adenomatous hyperplasia (Figure 4B), which are precancerous

lesions for squamous cell carcinoma and adenocarcinoma,

respectively. Neoplastic lesions, including squamous cell

carcinomas (Figure 4C) and adenocarcinomas (Figure 4D),

were also detected in the SM

+ NNK exposed groups.

These preneoplastic and neoplastic lesions were not detected

in the control group or the AOX group. Preneoplastic lesions

including squamous metaplasia, squamous dysplasia and

atypical adenomatous hyperplasia were observed in the lung

tissues of 10 of 12 (83%) SM

+ NNK ferrets, but only 7 of 14

(50%) ferrets treated with SM

+ NNK but supplemented with

AOX (Table III), which constituted a trend (33% decrease, P

¼

0.11) toward lower incidence of preneoplastic lesions in the

SM

+ NNK + AOX group, compared with the SM + NNK

group. Neoplastic lesions including squamous cell carcinoma,

adenocarcinoma

and

adenosquamous

carcinoma

were

observed in the lung tissues of 6 of 12 (50%) ferrets treated

with SM

+ NNK, but only 2 out of 14 (14%) ferrets treated with

SM

+ NNK + AOX (Table III), which showed a trend (36%

decrease, P

¼ 0.09) toward lower incidence of neoplastic

lesions in the AOX supplemented group, compared with the

SM

+ NNK group. However, it is important to note that neither

preneoplastic lesions nor neoplastic lesions were observed in

the lung tissue of ferrets in the control or AOX alone groups

after 6 months of intervention (Table III). In addition, there

were no significant differences in ferret body weight among the

treatment groups at the beginning, during or after the 6-month

experiment (data not shown).

Table II.Lung concentrations of BC, AT, AA and retinoids (RA, ROH, RP) in the four groups of ferrets after 6 months of treatment with smoke, NNK and

antioxidants (BC, AT, AA) in various combinations BC (pmol/g) AT (nmol/g) Reduced AA (mmol/g) Total AA (mmol/g) RA (pmol/g) ROH (nmol/g) RP (nmol/g) Control NDa 28.8 ± 5.7a 0.65 ± 0.08a 1.40 ± 0.26a 23.71 ± 3.54a 1.17 ± 0.23a 12.9 ± 2.7a SM+ NNK NDa 33.1 ± 6.9a 0.47 ± 0.07b 0.87 ± 0.09b 2.37 ± 0.23b 0.92 ± 0.20a 18.8 ± 3.5a AOX 8542 ± 1979b 64.5 ± 18.6b 0.73 ± 0.10a 1.20 ± 0.10a 42.27 ± 2.50c 1.15 ± 0.21a 9.9 ± 1.1a SM+ NNK + AOX 7834 ± 1546b 114.5 ± 24.0c 0.50 ± 0.09b 0.87 ± 0.14b 29.33 ± 2.24a 1.32 ± 0.31a 21.5 ± 5.7a

SM+ NNK, smoke-exposed plus NNK-treated; AOX, antioxidant supplemented (BC, AT, AA); SM + NNK + AOX, smoke-exposed plus NNK-treated

plus antioxidant supplemented; RA, retinoic acid; ROH, retinol; RP, retinyl palmitate.

a,b,cFor a given column, data not sharing a common superscript letter are statistically significantly different at P< 0.05. Values are expressed as means ±

(5)

Fig. 1.Expressions of total JNK, phospho-JNK, phospho-ERK, total ERK and PCNA protein levels were measured by western blot in lung tissue from four groups of ferrets after 6 months of treatment [Ctrl, control

(n¼ 9); SM + NNK, smoke-exposed plus NNK-treated (n ¼ 10); AOX,

antioxidant supplemented (BC, AT, AA) (n¼ 9); SM + NNK + AOX,

smoke-exposed plus NNK-treated plus antioxidants (BC, AT, AA) supplemented (n¼ 12)]. (A) Upper portions of each panel are

representative western blots in the same order as in the bar graph. The bar graph shows the intensity of the protein signal of phospho-JNK/total JNK, which was expressed by the relative values means ± SEM. Asterisk indicates that this value is significantly different from all others (P< 0.05). Fisher’s Least Significant Difference test was performed. The sizes were 46 kDa for JNK and phospho-JNK. (B) Upper portions of each panel are representative western blots in the same order as in the bar graph. The bar graph shows the intensity of the protein signal of phospho-ERK/total ERK, which was expressed by the relative values means ± SEM. Asterisk indicates that this value is significantly different from all others (P< 0.01). The sizes were 42 and 44 kDa for phospho-ERK and total ERK. (C) Upper portions of each panel are representative western blots in the same order as in the bar graph. The bar graph shows the intensity of the protein signal of PCNA was expressed by the relative values means ± SEM. Asterisk indicates that this value is significantly different from others (P< 0.05). The size was 34 kDa for PCNA.

Fig. 2.Expressions of total p53, phospho-p53 and Bax protein levels were measured by western blot in lung tissue from four groups of ferrets after 6 months of treatment [Ctrl, control (n¼ 9): SM + NNK,

smoke-exposed plus NNK-treated (n¼ 10); AOX, antioxidant supplemented

(BC, AT, AA) (n¼ 9); SM + NNK + AOX, smoke-exposed plus

NNK-treated plus antioxidants (BC, AT, AA) supplemented (n¼ 12)]. (A)

Upper portions of each panel are representative western blots in the same order as in the bar graph. The bar graph shows the intensity of the protein signal of total p53 was expressed by the relative values means ± SEM. Asterisk indicates that this value is significantly different from all others (P< 0.05). The size was 53 kDa for total p53. (B) Upper portions of each panel are representative western blots in the same order as in the bar graph. The bar graph shows the intensity of the protein signal of phospho-p53 at serine 15, which was expressed by the relative values means ± SEM. Asterisk indicates that this value is significantly different from all others (P< 0.01). (C) Upper portions of each panel are representative western blots in the same order as in the bar graph. The bar graph shows the intensity of the protein signal of Bax, which was expressed by the relative values means ± SEM. Asterisk indicates that this value is significantly different from others (P< 0.05). The size was 23 kDa for Bax.

(6)

Discussion

To our knowledge, this is the first report of an in vivo study

showing a beneficial effect of a combination of antioxidant

vitamins (BC, AT and AA) against lung damage induced by

tobacco smoke and NNK treatment in ferrets. The present

study shows that the ferret is a good model for studying

lung cancer chemoprevention with antioxidants and for

study-ing the molecular mechanism of carcinogenesis in the early

stages of smoke-related lung cancer.

Previously, we demonstrated the level of BC in the lung

tissue of smoke-exposed animals receiving low-dose BC

(0.48 mg/kg body wt/day) or high-dose BC (2.4 mg/kg

30 mg body wt/day) was significantly lowered with smoke

exposure (33). However, in the present study, the concentration

of BC was not lower with SM

+ NNK (Table II) when the

animal was fed with BC (0.85 mg/kg body wt/day) in the

presence of AT and AA, indicating that combined AT and

AA can prevent smoke-enhanced BC degradation. This is

in agreement with our previous demonstration that the

forma-tion of excentric cleavage metabolites of BC are higher in the

lungs of smoke-exposed ferrets than non-smoke-exposed

ferrets (6) and in vitro tissue incubation results showing that

this enhancement of excentric cleavage of BC can be blocked

by AT and AA (12).

We selected an AT dose of 22 mg/kg body wt, equivalent

to a daily intake of 100 mg AT in a 70 kg man based on a pilot

study revealing that the bioavailability of AT in the ferret

is

6% of that in a human. This dose has been shown in clinical

trials to improve immune responsiveness (40.2–536 mg AT

equivalents) (38) and to protect against LDL oxidation (17.4–

804 mg AT equivalents) without any toxic effect (39,40). For

the AA dosage, 3 mg/kg body wt/day in ferret (equivalent

to 210 mg AA in a 70 kg man) was chosen, since this dose

has been shown to afford smokers an equivalent protection

from AA hypovitaminosis as nonsmokers (41,42).

The lung AT concentration was significantly higher in the

AOX supplemented groups (Table II). Unexpectedly, however,

lung AT concentration was not lower in SM

+ NNK group

than in the control group. There is a lack of information about

the lung tissue concentration of AT in smokers and conflicting

information on the effects of smoke on plasma AT. Whereas

AT concentration was lowered by smoke in most in vitro

incubation studies (1,43,44), the plasma AT concentrations

of smokers either did not differ (44) or were lower than

those found in nonsmokers (45). However, we found that

AT concentration in lung tissue was higher with SM

+

NNK

+ AOX, compared with that of the control with AOX

supplementation. The mechanism for this is currently

unknown.

The concentration of lung AA was lower in the SM

+ NNK

group compared with the control groups in this study, in

concordance with previous human studies [(e.g. smokers

have significantly lower plasma levels of vitamin C compared

with nonsmokers) (46)]. This implies that the dose of smoke

exposure in this animal study was sufficient to produce a

smoking environment that mimics the human condition.

Inter-estingly, even with AA supplementation (3 mg/kg body wt,

which is equivalent to 210 mg/day in humans), lung levels of

AA in the smoke-exposed ferrets were not restored to control

levels (Table II). Therefore, it is possible that the function of

BC and AT in the lung tissue of smokers in the ATBC study

(2), and in a recent trial of head and neck cancer patients using

combined BC

+ AT to prevent second primary cancers (47),

was diminished by lack of sufficient AA to recycle both

antioxidants. The levels of reduced AA and total AA were

not different after AA supplementation in lung tissues of

ferrets, which is probably due to saturation of tissue AA,

since unlike humans, ferrets synthesize their own AA (48,49).

Cigarette smoke exposure is a strong risk factor for

lung cancer since it promotes genomic instability and the

development of neoplasia by modulating molecular pathways

involved in cell differentiation, cell proliferation and

apop-tosis. It has been reported that components of smoke or

smoke exposure itself increase MAPK, including JNK

(50–52) and ERK phosphorylation (52,53), in cell models.

In the present study, we demonstrated that phosphorylation

Fig. 3. Localization of phospho-p53 in the squamous carcinoma region in

the SM+ NNK group after 6 months of SM + NNK treatment. (A)

Non-tumor region and tumor region (· 100). (B) Non-tumor region (higher magnification, 400·) from (A); (C), Tumor-region (higher magnification, 400·) from (A).

(7)

of JNK and ERK was higher in the SM

+ NNK-treated ferrets,

as compared with the control group and this increased

phos-phorylation

was

inhibited

by

AOX

supplementation

(Figure 1A and B). It has been reported that activation and

phosphorylation of JNK at serine-63 and serine-73 residues

can result in activation of AP-1 (54,55), which may explain

increased cell proliferation (Figure 1C) and tumorigenesis

induced by smoke-exposure and NNK treatment. The levels

of total JNK and total ERK proteins were not different across

treatment groups, indicating that SM

+ NNK treatment and

AOX affect JNK and ERK activities by phosphorylation of

JNK and ERK, rather than by affecting protein amount.

Importantly, we have shown that lower levels of RA in

the lungs of the SM

+ NNK group were completely restored

to normal levels by AOX supplementation in our studies

(Tables I and II). Interestingly, we have previously shown

that high dose BC (2.4 mg/kg body wt/day) supplementation

decreased RA concentrations in the lungs of smoke-exposed

ferrets in vivo (6) and that the production of RA from BC

in the lung tissue of smoke exposed ferrets was substantially

increased by adding AT and AA to in vitro incubations (12).

This present in vivo study confirms our in vitro observations (8)

and indicates that combined AT and AA may enhance central

cleavage of BC, thereby increasing RA production from BC,

or inhibit excentric cleavage of BC, thereby decreasing of RA

catabolism. RA is the ligand for retinoid receptors (RARs and

RXRs) and appears to have the potential for chemopreventive

protection against cancer (56). It has been shown that RA

regulates the growth and differentiation of bronchial epithelial

cells in vitro and suppresses lung carcinogenesis in animal

studies (57). Recently, we demonstrate that 9-cis RA

supple-mentation in the A/J mouse model provides protection against

lung carcinogenesis and this effect may be mediated in

part by 9-cis RA induction of RAR-b (58). Although the

effi-cacy and complex biological functions of retinoids in human

lung cancer prevention need more investigation (59), our study

indicates that the restoration of normal levels of RA by AOX

supplementation in smoke-exposed lung tissue may contribute

to the protective effects of AOX against lung carcinogenesis. It

has been reported that RA can inhibit phosphorylation

of MAPKs, such as JNK and ERK, by upregulation of MAP

kinase-phosphatase-1 which dephosphorylates MAPK (60–

62), thereby preventing abnormal cell proliferation. This

sug-gests that the mechanism behind the inhibitory effect of AOX

supplementation against the phosphorylation of JNK and ERK

could be due to increased lung RA levels in SM

+ NNK + AOX

group. This is supported by our recent observation that the

induction of MAP kinase-phosphatase-1 was associated with

the concentration of RA in the lungs of ferrets (25). This was

also supported by the fact that PCNA was significantly higher

in the lungs of SM

+ NNK when the levels of RA were lower as

Fig. 4.Representative preneoplastic and neoplastic lesions seen in the lungs of ferrets after 6 months of SM+ NNK treatment. (A) Squamous dysplasia, HE (400·). (B) Atypical adenomatous hyperplasia, HE (100·). (C) Squamous cell carcinoma, HE (200·). (D) Adenocarcinoma, HE (200·).

Table III. The incidence of preneoplastic lesion and neoplastic lesions in the four groups of ferrets after 6 months of treatment

Preneoplastic lesion1,3 Neoplastic lesion2,3

Control 0/9 (0%)a 0/9 (0%)a AOX 0/9 (0%)a 0/9 (0%)a SM+ NNK 10/12 (83%)b P¼ 0.11 6/12 (50%) b P¼ 0.09 SM+ NNK + AOX 7/14 (50%)b 2/14 (14%)a,b

SM+ NNK, smoke-exposed plus NNK-treated; AOX, antioxidant

supplemented (b-carotene, a-tocopherol and ascorbic acid);

SM+ NNK + AOX, smoke-exposed plus NNK-treated plus

antioxidant supplemented.

1Preneoplastic lesion, number of ferrets with preneoplastic lesions including squamous metaplasia, squamous dysplasia and atypical adenomatous hyperplasia/the total number of ferrets in each group (%). 2

Neoplastic lesion, number of ferrets with neoplastic lesions including squamous cell carcinoma, adenocarcinoma and adenosquamous carcinoma/the total number of ferrets in each group (%). 3

Data not sharing a common superscript letters for a given value indicate that the incidence of lung squamous metaplasia statistically significantly different from each other (p< 0.05, Fisher’s Exact test).

(8)

compared with the control group or the AOX supplemented

group (Figure 1C). Furthermore, this increase in PCNA

labeling was prevented when the lower levels of lung RA in

the SM

+ NNK group were restored to normal in the SM +

NNK

+ AOX group. In addition, our study suggests that the

inhibition of JNK activation by the combined antioxidants may

help to ‘rescue’ the functions of RAR because it has been

recently reported that activation of JNK contributes to RAR

dysfunction by phosphorylating RAR-alpha and inducing

degradation through the ubiquitin-proteasomal pathway (63).

This hypothesis warrants further investigation.

Phosphorylation of JNK and ERK mediate phosphorylation

of p53, an important tumor suppressor that plays a critical

role in the cell-injury response to various stressors (19,20).

Overexpression of p53 protein is associated with cigarette

consumption (64). There is a dose–response relationship

between the quantity of tobacco consumed and the frequency

of p53 gene mutation in lung cancer patients (65). p53

phosphorylation at serine-15 facilitates both the accumulation

and functional activation of p53 (66,67). Solhaug et al. (52)

have shown that B[a]P-metabolites, B[a]P-7,8-DHD and

BPDE-1 induced accumulation and phosphorylation of p53

at serine-15 in Hepa1c1c7 cells. In the present study, we

have shown combined AOX (BC, AT and AA) inhibited the

phosphorylation of p53 induced by SM

+ NNK treatment

(Figure 2B). These findings are consistent with the lower

expression of phospho-JNK and phospho-ERK in the SM

+

NNK

+ AOX groups as compared with the SM + NNK group.

In addition, total p53 protein level was also increased in

parallel with phospho-p53 in SM

+ NNK-treated groups versus

all other groups, suggesting that the accumulation and

func-tional activation of p53 by its phosphorylation are mediated by

JNK and ERK.

p53 phosphorylation at serine-15 was upregulated in the

SM

+ NNK groups compared with the control group

(Figure 2B). Post-translational modifications of p53 by

phos-phorylation have been implicated in its stabilization and

transcriptional activation (68). In response to stress, p53

undergoes rapid phosphorylation on several residues,

includ-ing serine-6, 9, 20, 37, 46, 81, 389 and 392. At later time points

following DNA damage, p53 is phosphorylated on residues 15

and 372 (69). Although we showed higher phospho-p53

expression in the SM

+ NNK group than in the control

group, there are no reports of higher p53 phosphorylation

in the tumor regions compared with the non-tumor regions.

Therefore, localization of p53 phosphorylation (serine-15)

was studied using immunohistochemistry in tumor regions

(Figure 3) in order to examine whether the increased

phos-phorylation of p53 in tumor regions may contribute to the

higher levels of phospho-p53 seen in the SM

+ NNK group

(Figure 2B). This change in phosphorylation may regulate

downstream events such as apoptosis by activation of Bax

in the tumor progress. Our findings point to a general shift

in the regulation of p53 phosphorylation, which takes place

during tumor development and/or progression.

As one of the downstream transcriptional targets of p53,

Bax is a Bcl-2 related protein promoting apoptosis. Bax has

been shown to contain p53-binding sites in its promoter and is

upregulated in response to DNA damage (70). The level of Bax

was higher in SM

+ NNK group compared with that of the

control group. Furthermore, AOX supplementation (SM

+

NNK

+

AOX)

prevented

the

upregulation

of

Bax

induced by SM

+ NNK (Figure 2C). This result indicates

that SM

+ NNK treatment may induce apoptosis as well as

proliferation. Since normal cells must maintain a homeostatic

balance between cell proliferation and apoptosis, the loss of

this balance may contribute to lung carcinogenesis. This would

particularly be the case if abnormal cell proliferation increased

much more than apoptosis, even if both were increased by

smoke exposure and NNK treatment.

In this study, we showed that BC (equivalent to 12 mg/day in

humans) in the presence of AT and AA prevented both

preneoplastic lesions and neoplasia induced by 6 months of

tobacco smoke exposure and NNK treatment (Table III).

Although this effect on lung lesions is not statistically

signi-ficant due to the small sample size, our biochemical and

molecular mechanistic analyses as well as our short-term

study data (Table I) fully support the beneficial effects of

AOX against lung carcinogenesis. Interestingly, in contrast

to our previous observation that lung squamous metaplastic

lesions were increased in ferrets exposed to high dose BC

(equivalent to 30 mg/day in humans) (6), the same dose of

BC supplementation in the presence of AT and AA in

smoke-exposed ferrets reduced the number of ferrets having lung

squamous metaplasia, as compared with the unsupplemented

smoke exposed group (1/9 versus 4/9, Table I). In addition, we

showed that a low dose of BC (equivalent to 6 mg/day in

humans) in the presence of AT and AA had a significant

pro-tective effect on lung squamous metaplasia induced by tobacco

smoke (Table I). However, we only observed a trend for the

protection of AOX supplementation against lung lesions in the

ferrets exposed to both tobacco smoke and NNK (Table III). A

possible explanation for the difference between these two

studies is that the ferrets were only exposed to smoke in the

short-term study and only squamous metaplasia was detected,

whereas the combined smoke-exposure and NNK injection

were used in the long-term study and preneoplastic lesions

including squamous metaplasia, squamous dysplasia and

atyp-ical adenomatous hyperplasia were detected. Therefore, the

NNK injection in addition to smoke-exposure in the ferrets

may have overwhelmed the ability of antioxidants to provide

strong protection. Another possible explanation is that the

AOX supplementation prevented against the cancer promoting

effects of smoke-exposure, such as smoke-induced

inflamma-tion and free radical damage, while not offering protecinflamma-tion

against NNK chemically induced lung carcinogenesis. The

investigation as to whether antioxidants can protect against

lung cancer induced by NNK alone is currently underway

in this laboratory.

In summary, this in vivo study showed that vitamin E and

vitamin C acting together play an important role in terms

of inhibiting oxidation of BC and facilitating conversion of

BC into RA in smoke-exposed lung tissue. AOX

supplementa-tion may exert its protective effect against lung carcinogenesis

by maintaining normal concentrations of RA and inhibiting

phosphorylation of JNK, ERK and p53. These studies and

the known biochemical interactions of BC, vitamin E and

vitamin C suggest that this combination of nutrients, rather

than individual agents, could be an effective chemopreventive

strategy against lung cancer in smokers.

Acknowledgements

Y.K. was supported by NIH training grant T32 DK62032-11. The authors thank both Cognis, Corp. and DSM Corp. for funding our pilot study and Heather Mernitz for help in the preparation of the manuscript. Supported by the

(9)

NIH grant R01CA49195 and U.S. Department of Agriculture, under agreement NO. 1950-51000-064. Any opinions, findings, conclusion or recommendations expressed in this publication are those of the author(s) and do not necessarily reflects the views of the U.S. Department of Agriculture.

Conflict of Interest Statement: None declared.

References

1. Mayne,S.T., Handelman,G.J. and Beecher,G. (1996) Beta-carotene and lung cancer promotion in heavy smokers—a plausible relationship? J. Natl Cancer Inst., 88, 1513–1515.

2. The Alpha-Tocopherol, Beta Carotene Cancer Prevention Study Group. (1994) The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers. N. Engl. J. Med, 330, 1029–1035.

3. Omenn,G.S., Goodman,G.E., Thornquist,M.D. et al. (1996) Risk factors for lung cancer and for intervention effects in CARET, the Beta-Carotene and Retinol Efficacy Trial. J. Natl Cancer Inst., 88, 1550–1559.

4. Wang,X.D. (2004) Carotenoid Oxidative/Degradative Products and Their Biological Activities. Marcel Dekker, Inc., New York, NY.

5. Wang,X.D. (2004) Mechanistic Understanding of Potential Adverse Effects of Beta-Carotene Supplementation. Wiley-VCH, Weinheim.

6. Wang,X.D., Liu,C., Bronson,R.T., Smith,D.E., Krinsky,N.I. and

Russell,M. (1999) Retinoid signaling and activator protein-1 expression in ferrets given beta-carotene supplements and exposed to tobacco smoke. J. Natl Cancer Inst., 91, 60–66.

7. Salgo,M.G., Cueto,R., Winston,G.W. and Pryor,W.A. (1999) Beta carotene and its oxidation products have different effects on microsome mediated binding of benzo[a]pyrene to DNA. Free Radic. Biol. Med., 26, 162–173.

8. Liu,C., Russell,R.M. and Wang,X.D. (2003) Exposing ferrets to cigarette smoke and a pharmacological dose of beta-carotene supplementation enhance in vitro retinoic acid catabolism in lungs via induction of cytochrome P450 enzymes. J. Nutr., 133, 173–179.

9. Bohm,F., Edge,R., McGarvey,D.J. and Truscott,T.G. (1998) Beta-carotene with vitamins E and C offers synergistic cell protection against NOx. FEBS Lett., 436, 387–389.

10. Bohm,F. (1997) Cartenoids enhance vitamin E antioxidant efficiency. J. Am. Chem. Soc., 119, 621–622.

11. Weitberg,A.B. and Corvese,D. (1997) Effect of vitamin E and beta-carotene on DNA strand breakage induced by tobacco-specific nitrosamines and stimulated human phagocytes. J. Exp. Clin. Cancer Res., 16, 11–14. 12. Liu,C., Russell,R.M. and Wang,X.D. (2004) Alpha-tocopherol and ascorbic

acid decrease the production of beta-apo-carotenals and increase the formation of retinoids from beta-carotene in the lung tissues of cigarette smoke-exposed ferrets in vitro. J. Nutr., 134, 426–430.

13. Chan,A.C. (1993) Partners in defense, vitamin E and vitamin C. Can. J. Physiol. Pharmacol., 71, 725–731.

14. Black,H.S. (1998) Radical interception by carotenoids and effects on UV carcinogenesis. Nutr. Cancer, 31, 212–217.

15. Yong,L.C., Brown,C.C., Schatzkin,A., Dresser,C.M., Slesinski,M.J., Cox,C.S. and Taylor,P.R. (1997) Intake of vitamins E, C, and A and risk of lung cancer. The NHANES I epidemiologic followup study. First National Health and Nutrition Examination Survey. Am. J. Epidemiol., 146, 231–243.

16. Wright,M.E., Mayne,S.T., Stolzenberg-Solomon,R.Z., Li,Z., Pietinen,P., Taylor,P.R., Virtamo,J. and Albanes,D. (2004) Development of a compre-hensive dietary antioxidant index and application to lung cancer risk in a cohort of male smokers. Am. J. Epidemiol., 160, 68–76.

17. Davis,R.J. (2000) Signal transduction by the JNK group of MAP kinases. Cell, 103, 239–252.

18. Rincon,M., Flavell,R.A. and Davis,R.A. (2000) The JNK and P38 MAP kinase signaling pathways in T cell-mediated immune responses. Free Radic. Biol. Med., 28, 1328–1337.

19. She,Q.B., Chen,N. and Dong,Z. (2000) ERKs and p38 kinase phosphorylate p53 protein at serine 15 in response to UV radiation. J. Biol. Chem., 275, 20444–20449.

20. Smeal,T., Binetruy,B., Mercola,D.A., Birrer,M. and Karin,M. (1991)

Oncogenic and transcriptional cooperation with Ha-Ras requires

phosphorylation of c-Jun on serines 63 and 73. Nature, 354, 494–496. 21. Karin,M., Liu,Z. and Zandi,E. (1997) AP-1 function and regulation.

Curr. Opin. Cell Biol., 9, 240–246.

22. Gupta,P. and Prywes,R. (2002) ATF1 phosphorylation by the ERK MAPK pathway is required for epidermal growth factor-induced c-jun expression. J. Biol. Chem., 277, 50550–50556.

23. Yeh,P.Y., Chuang,S.E., Yeh,K.H., Song,Y.C., Chang,L.L. and Cheng,A.L. (2004) Phosphorylation of p53 on Thr55 by ERK2 is necessary for doxorubicin-induced p53 activation and cell death. Oncogene, 23, 3580–3588.

24. Siliciano,J.D., Canman,C.E., Taya,Y., Sakaguchi,K., Appella,E. and Kastan,M.B. (1997) DNA damage induces phosphorylation of the amino terminus of p53. Genes Dev., 11, 3471–3481.

25. Liu,C., Russell,R.M. and Wang,X.D. (2004) Low dose beta-carotene supplementation of ferrets attenuates smoke-induced lung phosphorylation of JNK, p38 MAPK, and p53 proteins. J. Nutr., 134, 2705–2710. 26. Wang,X.D., Krinsky,N.I., Marini,R.P., Tang,G., Yu,J., Hurley,R., Fox,J.G.

and Russell,R.M. (1992) Intestinal uptake and lymphatic absorption of beta-carotene in ferrets: a model for human beta-carotene metabolism. Am. J. Physiol., 263, G480–486.

27. Gugger,E.T., Bierer,T.L., Henze,T.M., White,W.S. and Erdman,J.W Jr (1992) Beta-carotene uptake and tissue distribution in ferrets (Mustela putorius furo). J. Nutr., 122, 115–119.

28. Wang,X.D., Tang,G.W., Fox,J.G., Krinsky,N.I. and Russell,R.M. (1991) Enzymatic conversion of beta-carotene into beta-apo-carotenals and retin-oids by human, monkey, ferret, and rat tissues. Arch. Biochem. Biophys., 285, 8–16.

29. Wang,X.D., Russell,R.M., Marini,R.P., Tang,G., Dolnikowski,G.G., Fox,J.G. and Krinsky,N.I. (1993) Intestinal perfusion of beta-carotene in the ferret raises retinoic acid level in portal blood. Biochim. Biophys. Acta, 1167, 159–164.

30. Wang,X.D., Russell,R.M., Liu,C., Stickel,F., Smith,D.E. and Krinsky,N.I. (1996) Beta-oxidation in rabbit liver in vitro and in the perfused ferret liver contributes to retinoic acid biosynthesis from beta-apocarotenoic acids. J. Biol. Chem., 271, 26490–26498.

31. Kim,Y., Liu,X.S., Liu,C., Smith,D.E., Russell,R.M. and Wang,X.-D. (2005) Induction of pulmonary neoplasia in the smoke-exposed ferret by 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK): a model for human lung cancer. Cancer Lett., 217, 1–17.

32. Koppang,N., Rivenson,A., Dahle,H.K. and Hoffmann,D. (1997) A study of tobacco carcinogenesis, LIII: carcinogenicity of N0-nitrosonornicotine (NNN) and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) in mink (Mustela vison). Cancer Lett., 111, 167–171.

33. Liu,C., Wang,X.D., Bronson,R.T., Smith,D.E., Krinsky,N.I. and

Russell,R.M. (2000) Effects of physiological versus pharmacological beta-carotene supplementation on cell proliferation and histopathological changes in the lungs of cigarette smoke-exposed ferrets. Carcinogenesis, 21, 2245–2253.

34. Yeum,K.J., Taylor,A., Tang,G. and Russell,R.M. (1995) Measurement of carotenoids, retinoids, and tocopherols in human lenses. Invest Ophthalmol. Vis. Sci., 36, 2756–2761.

35. McKay,D.L., Perrone,G., Rasmussen,H., Dallal,G., Hartman,W., Cao,G., Prior,R.L., Roubenoff,R. and Blumberg,J.B. (2000) The effects of a multivitamin/mineral supplement on micronutrient status, antioxidant capacity and cytokine production in healthy older adults consuming a fortified diet. J. Am. Coll. Nutr., 19, 613–621.

36. Travis,W.D. (2002) Pathology of lung cancer. Clin. Chest Med., 23, 65–81, viii.

37. Dungworth,D.L., Hauser,B., Hahn,F.F., Wilson,D.W., Taenichen,T. and Harkema,J.R. (1999) Histological Classification of Tumors of the Respir-atory System of Domestic Animals. The Armed Forces Institue of Patho-logy, Washington, DC.

38. Meydani,S.N., Meydani,M., Blumberg,J.B., Leka,L.S., Siber,G.,

Loszewski,R., Thompson,C., Pedrosa,M.C., Diamond,R.D. and

Stollar,B.D. (1997) Vitamin E supplementation and in vivo immune response in healthy elderly subjects. A randomized controlled trial. JAMA, 277, 1380–1386.

39. Jialal,I., Fuller,C.J. and Huet,B.A. (1995) The effect of alpha-tocopherol supplementation on LDL oxidation. A dose–response study. Arterioscler. Thromb. Vasc. Biol., 15, 190–198.

40. Princen,H.M., van Duyvenvoorde,W., Buytenhek,R., van der Laarse,A., van Poppel,G., Gevers Leuven,J.A. and van Hinsbergh,V.W. (1995) Supplementation with low doses of vitamin E protects LDL from lipid peroxidation in men and women. Arterioscler. Thromb. Vasc. Biol., 15, 325–333.

41. Schectman,G. (1993) Estimating ascorbic acid requirements for cigarette smokers. Ann. N. Y. Acad. Sci., 686, 335–345; discussion 345–346.

42. Mooney,L.A., Madsen,A.M., Tang,D., Orjuela,M.A., Tsai,W.Y.,

Garduno,E.R. and Perera,F.P. (2005) Antioxidant vitamin

supple-mentation reduces benzo(a)pyrene-DNA adducts and potential

cancer risk in female smokers. Cancer Epidemiol. Biomarkers Prev., 14, 237–242.

(10)

43. Frei,B., Stocker,R. and Ames,B.N. (1988) Antioxidant defenses and lipid peroxidation in human blood plasma. Proc. Natl Acad. Sci. USA, 85, 9748–9752.

44. Leonard,S.W., Bruno,R.S., Paterson,E., Schock,B.C., Atkinson,J.,

Bray,T.M., Cross,C.E. and Traber,M.G. (2003) 5-nitro-gamma-tocopherol increases in human plasma exposed to cigarette smoke in vitro and in vivo. Free Radic. Biol. Med., 35, 1560–1567.

45. Bolton-Smith,C., Casey,C.E., Gey,K.F., Smith,W.C. and Tunstall-Pedoe,H. (1991) Antioxidant vitamin intakes assessed using a food-frequency ques-tionnaire: correlation with biochemical status in smokers and non-smokers. Br. J. Nutr., 65, 337–346.

46. Schectman,G., Byrd,J.C. and Gruchow,H.W. (1989) The influence of smoking on vitamin C status in adults. Am. J. Public Health, 79, 158–162. 47. Bairati,I., Meyer,F., Gelinas,M. et al. (2005) A randomized trial of anti-oxidant vitamins to prevent second primary cancers in head and neck cancer patients. J. Natl Cancer Inst., 97, 481–488.

48. McLain,D.E., Thomas,J.A. and Fox,J.G. (1988) Nutrition. Lea & Febiger, Philadelphia.

49. Fiala,E.S., Sohn,O.S., Wang,C.X. et al. (2005) Induction of preneoplastic lung lesions in guinea pigs by cigarette smoke inhalation and their exacer-bation by high dietary levels of vitamins C and E. Carcinogenesis, 26, 605–612.

50. Lei,W., Yu,R., Mandlekar,S. and Kong,A.N. (1998) Induction of apoptosis and activation of interleukin 1beta-converting enzyme/Ced-3 protease (caspase-3) and c-Jun NH2-terminal kinase 1 by benzo(a)pyrene. Cancer Res., 58, 2102–2106.

51. Yoshii,S., Tanaka,M., Otsuki,Y., Fujiyama,T., Kataoka,H., Arai,H., Hanai,H. and Sugimura,H. (2001) Involvement of alpha-PAK-interacting exchange factor in the PAK1-c-Jun NH(2)-terminal kinase 1 activation and apoptosis induced by benzo[a]pyrene. Mol. Cell. Biol., 21, 6796–6807. 52. Solhaug,A., Ovrebo,S., Mollerup,S., Lag,M., Schwarze,P.E., Nesnow,S.

and Holme,J.A. (2005) Role of cell signaling in B[a]P-induced apop-tosis: characterization of unspecific effects of cell signaling inhibitors and apoptotic effects of B[a]P metabolites. Chem. Biol. Interact., 151, 101–119.

53. Yang,Y., Zhang,Z., Mukherjee,A.B. and Linnoila,R.I. (2004) Increased susceptibility of mice lacking Clara cell 10-kDa protein to lung tumori-genesis by 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone, a potent carcinogen in cigarette smoke. J. Biol. Chem., 279, 29336–29340. 54. Whitmarsh,A.J. and Davis,R.J. (1996) Transcription factor AP-1 regulation

by mitogen-activated protein kinase signal transduction pathways. J. Mol. Med., 74, 589–607.

55. Minden,A. and Karin,M. (1997) Regulation and function of the JNK subgroup of MAP kinases. Biochim. Biophys. Acta, 1333, F85–104. 56. Altucci,L. and Gronemeyer,H. (2001) The promise of retinoids to fight

against cancer. Nat. Rev. Cancer, 1, 181–193.

57. Lippman,S.M. and Hong,W.K. (1992) Retinoid Chemoprevention of Upper Aerodigestive Tract Carcinogenesis. Lippincott, Philadelphia.

58. Mernitz,H., Smith,D.E., Zhu,A.X. and Wang,X.D. (2006) 9-Cis reginoic acid inhibits lung carcinogenesis in the A/J mouse model by inducing RAR-beta expression but not affecting cyclooxygenase-2 protein. Cancer Lett., (in press).

59. Khuri,F.R. and Lotan,R. (2004) Retinoids in lung cancer: friend, foe, or fellow traveler? J. Clin. Oncol., 22, 3435–3437.

60. Hirsch,D.D. and Stork,P.J. (1997) Mitogen-activated protein kinase phosphatases inactivate stress-activated protein kinase pathways in vivo. J. Biol. Chem., 272, 4568–4575.

61. Lee,H.Y., Sueoka,N., Hong,W.K., Mangelsdorf,D.J., Claret,F.X. and Kurie,J.M. (1999) All-trans-retinoic acid inhibits Jun N-terminal kinase by increasing dual-specificity phosphatase activity. Mol. Cell. Biol., 19, 1973–1980.

62. Chung,J., Chavez,P.R., Russell,R.M. and Wang,X.D. (2002) Retinoic acid inhibits hepatic Jun N-terminal kinase-dependent signaling pathway in ethanol-fed rats. Oncogene, 21, 1539–1547.

63. Srinivas,H., Juroske,D.M., Kalyankrishna,S., Cody,D.D., Price,R.E., Xu,X.C., Narayanan,R., Weigel,N.L. and Kurie,J.M. (2005) c-Jun N-terminal kinase contributes to aberrant retinoid signaling in lung cancer cells by phosphorylating and inducing proteasomal degradation of retinoic acid receptor alpha. Mol. Cell. Biol., 25, 1054–1069.

64. Gosney,J.R., Butt,S.A., Gosney,M.A. and Field,J.K. (1993) Exposure to cigarette smoke and expression of the protein encoded by the p53 gene in bronchial carcinoma. Ann. N. Y. Acad. Sci., 686, 243–247; discussion 247–248.

65. Kondo,K., Tsuzuki,H., Sasa,M., Sumitomo,M., Uyama,T. and Monden,Y. (1996) A dose-response relationship between the frequency of p53 muta-tions and tobacco consumption in lung cancer patients. J. Surg. Oncol., 61, 20–26.

66. Kwon,Y.W., Ueda,S., Ueno,M., Yodoi,J. and Masutani,H. (2002) Mech-anism of p53-dependent apoptosis induced by 3-methylcholanthrene: involvement of p53 phosphorylation and p38 MAPK. J. Biol. Chem., 277, 1837–1844.

67. Shieh,S.Y., Ikeda,M., Taya,Y. and Prives,C. (1997) DNA

damage-induced phosphorylation of p53 alleviates inhibition by MDM2. Cell, 91, 325–334.

68. Meek,D.W. (1998) Multisite phosphorylation and the integration of stress signals at p53. Cell. Signal, 10, 159–166.

69. Buschmann,T., Adler,V., Matusevich,E., Fuchs,S.Y. and Ronai,Z. (2000) p53 phosphorylation and association with murine double minute 2,

c-Jun NH2-terminal kinase, p14ARF, and p300/CBP during the cell

cycle and after exposure to ultraviolet irradiationCancer Res., 60, 896–900.

70. Miyashita,T. and Reed,J.C. (1995) Tumor suppressor p53 is a

direct transcriptional activator of the human bax gene. Cell, 80, 293–299.

Received August 24, 2005; revised December 18, 2005; accepted January 3, 2006

Cytaty

Powiązane dokumenty

Key words: intermittent hypoxic training, ultra-structural lung and heart tissues changes, biological barriers thickness, mitochondria,

The aim of our study was the immunohistoche- mical evaluation of p53, bcl-2 and nm23 expression in squamous cell lung cancers in relation to basic cli- nical and pathological

W polskich rekomendacjach dotyczących le- czenia raka płuca zaleca się u chorych starszych, którzy pozostają w dobrym stanie ogólnym, stoso- wanie tych samych zasad i metod leczenia,

Zespół wyniszczenia nowotworowego charak- teryzuje się zmniejszeniem masy ciała (dotyczącym głównie tkanki mięśniowej i tłuszczowej), zaburze- niami łaknienia,

The right lower lobe resection has proba- bly been sufficient treatment for both lung cancer and organising pneumonia.. Having analyzed the literature, no similar co-existence of

Na przestrzeni lat zaobserwowano, zarówno u mężczyzn, jak i u kobiet, stały wzrost liczby chorych na gruczolakoraka, a obniżanie się liczby chorych na raka płaskonabłonko- wego i

The intratumoral expression of vascular endothelial growth factor and interleukin-8 associated with angiogenesis in non-small cell lung carcinoma patients. Inhibitory neoangiogenezy

— The treatment of choice in patients with locally advanced non-small-cell lung cancer is radical chemoradiation or — in the case of contraindica- tions to chemotherapy —