Sie sind auf Seite 1von 11

Available online at www.sciencedirect.

com

Chemico-Biological Interactions 173 (2008) 32–42

Protective effects of Ginkgo biloba against rat liver carcinogenesis


Marcos C. Dias a , Maria A.M. Rodrigues b , Maria C.H. Reimberg c , Luı́s F. Barbisan a,∗
a UNESP São Paulo State University, Institute of Biosciences, Department of Morphology, Botucatu, SP 18618-000, Brazil
b UNESP São Paulo State University, Faculty of Medicine, Department of Pathology, Botucatu, SP 18618-000, Brazil
c CentroFlora Group, Botucatu, SP 18603-970, Brazil

Received 7 November 2007; received in revised form 11 January 2008; accepted 21 January 2008
Available online 12 February 2008

Abstract
Ginkgo biloba (EGb) has been proposed as a promising candidate for cancer chemoprevention and has shown protective effects
on the liver against chemically induced oxidative injury and fibrosis. The potential beneficial effects of EGb were investigated in
two rat liver carcinogenesis bioassays induced by diethylnitrosamine (DEN). In a short-term study for anti-initiating screening,
male Wistar rats were fed a basal diet or supplemented diet with 500 or 1000 ppm EGb and initiated 14 days later with a single dose
of DEN (100 mg/kg i.p.). The respective groups were killed 24 h or 2 weeks after DEN-initiation. Liver samples were collected for
the analysis of proliferating cell nuclear antigen (PCNA), transforming growth factor alpha (TGF-␣), p53, apoptosis and induction
of single hepatocytes and minifoci positive for the enzyme glutathione S-transferase P-form (GST-P). In a medium-term study for
anti-promoting screening, the animals received a single dose of DEN (200 mg/kg i.p.) and, 2 weeks later, were fed a basal diet or
supplemented diet with 500 or 1000 ppm EGb for 6 weeks. All animals underwent 70% partial hepatectomy (PH) at week 3 and
killed at week 8. Liver samples were colleted to analyze development of preneoplastic foci of altered hepatocytes (FAH) expressing
GST-P. In the short-term study, pretreatment of rats with 1000 ppm EGb significantly reduced the rates of cell proliferation, apoptosis
and p53, TGF-␣ immunoreactivity and the number of GST-P-positive hepatocytes. In the medium-term study, EGb treatment during
the post-initiation stage failed to reduce the development of DEN-induced GST-P-positive foci. Thus, EGb presented inhibitory
actions during initiation but not promotion of rat liver carcinogenesis induced by DEN.
© 2008 Elsevier Ireland Ltd. All rights reserved.

Keywords: Diethylnitrosamine; Putative initiated hepatocytes; GST-P-positive preneoplastic foci; Liver carcinogenesis; Ginkgo biloba; Chemopre-
vention

1. Introduction that has presented various pharmacological activities


[2–4]. This extract has shown several in vivo effects,
Ginkgo biloba extract (EGb) is a commercial medic- including augmentation of blood flow and inhibi-
inal herb which comes from the green leaves of the tion of platelet activating factor. It protects the cell
ginkgo tree, one of the oldest living plant species [1]. membrane against damage induced by free radicals
EGb is a mixture, composed mainly of flavone gly- and presents protective effects against myocardial and
cosides and terpenoids (ginkgolides and bilobalide), brain ischemia/reperfusion injury [2–5]. EGb has been
widely used in the clinical treatment of cardiovascular
and neurological diseases like Alzheimer’s, dementia,
∗ Corresponding author at: Departamento de Morfologia, Instituto labyrintopathies and other cognitive dysfunctions [2,6].
de Biociências, Universidade Estadual Paulista (UNESP), Botucatu,
18618-000 SP, Brazil. Tel.: +55 14 68116264; fax: +55 14 68116264.
EGb has shown protective effects on the liver against
E-mail address: barbisan@ibb.unesp.br (L.F. Barbisan). chemically induced oxidative injury and fibrosis [7–10].

0009-2797/$ – see front matter © 2008 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.cbi.2008.01.012
M.C. Dias et al. / Chemico-Biological Interactions 173 (2008) 32–42 33

Also, EGb has been shown to modulate the hepatic were kept in polypropylene cages (five animals/cage)
phase I enzymes, including the induction or inhibi- covered with metallic grids in a room maintained at
tion of specific cytochrome P450 (CYP) isozymes, and 22 ± 2 ◦ C, 55 ± 10% humidity under a 12-h light:12-h
phase II enzymes, specifically the induction of glutathion dark cycle. They were fed commercial NUVILAB-CR-
S-transferases, DT-diaphorase and quinine reductase 1 chow (NUVITAL, Curitiba, PR, Brazil) and water ad
[11–14]. Thus, changes in metabolizing phase I and libitum for a 2-week acclimation period before begin-
phase II enzymes by EGb can alter the balance between ning the experiment. Samples of lyophilized extract of
the activation of procarcinogens or detoxification of G. biloba leaves (EGb), generously supplied by the Cen-
potential carcinogens. troFlora Group (Botucatu, SP, Brazil), were obtained
There are few in vivo and in vitro studies on the from hydroalcoholic extraction by a spray dryer system.
potential anti-carcinogenic effects of EGb. Crude EGb The EGb (cod. 500821) used in this study contained
or their specific compounds induced cellular death or known amounts of approximately 24% flavone glyco-
anti-proliferative activities against the following cancer sides (i.e., quercetin, kaempferol and isorhamnetin) and
cell lines: HepG2, Hep 3B, and SMMC-7721 human 6% terpene trilactones (i.e., ginkgolide A–C, bilobalide)
hepatocellular [15,16], SCC 1483 human oral [17] and as determined by the HPLC method. EGb was supple-
OVCA429, 433 and 420 human ovarian [18]. EGb, when mented to basal diet at 500 and 1000 ppm, levels which
fed orally to female Swiss mice, reduced forestomach correspond to 1 and 2 times the doses that were used in
tumor multiplicity induced by benzo(a)pyrene [19]. EGb an in vivo feeding study for beneficial effects against rat
and bilobalide, given orally to male Fischer 344 rats, colon carcinogenesis [20].
inhibited the development of colonic aberrant crypt foci
induced by azoxymethane [20]. 2.2. Experimental design
Foci of altered hepatocytes (FAH) have been
described as putative preneoplastic lesions detected in The protocols used were consistent with Ethical Prin-
various rodent models of chemical liver carcinogenesis ciples for Animal Research adopted by the Brazilian
[21]. More recently, different types of FAH with similar College of Animal Experimentation (COBEA). The
morphological and biochemical alterations in the hepato- animals were randomly allocated to two experimental
cellular phenotype were identified in chronic human liver protocols: a short- and a medium-term liver bioassay,
diseases associated with, or predisposing to hepatocel- respectively (Fig. 1).
lular carcinoma [22]. Glutathione S-transferase P-form
(GST-P) expression is a useful marker for preneoplastic 2.2.1. Experiment 1 (short-term bioassay)
and neoplastic rat liver lesions [23]. Single hepatocytes This study was performed to investigate the modify-
and minifoci highly positive for GST-P develop very ing effects of EGb intake on the first stage of rat liver
early in carcinogen-treated rat liver, and are considered carcinogenesis in an initiation bioassay model that uses
precursors of large FAH and nodules [24,25]. The detec- the detection of initiated hepatocytes and minifoci pos-
tion of GST-P-positive single cells and minifoci or large itive for GST-P as the endpoint [24,25]. The animals
GST-P-positive FAH is an important tool for analyz- were randomly allocated into four groups: groups G1A
ing relevant carcinogenic or anti-carcinogenic responses and G4A were fed basal diet and groups G2A and G3A
during the initiation and promotion stages of rat liver were fed a diet supplemented with 500 or 1000 ppm EGb
carcinogenesis [26–28]. during 2 weeks. Then, groups G1A to G3A were given
Using a short-term (anti-initiating screening) and a a single i.p. injection of 100 mg/kg b.w. of diethylni-
medium-term (anti-promoting screening) liver bioassay, trosamine and group 4A (control) received 0.9% NaCl
the present study investigated the modifying influence (DEN vehicle). Twenty-four hours after DEN admin-
of a G. biloba extract (EGb) on the initiation and pro- istration, groups G1A to G3A were fed basal diet ad
moting phases of rat liver carcinogenesis induced by libitum. The animals were sacrificed 24 h or 2 weeks
diethylnitrosamine (DEN). after DEN treatment (end of week 4).

2. Material and methods 2.2.2. Experiment 2 (medium-term bioassay)


This study was performed to investigate the modify-
2.1. Animals and treatments ing effects of EGb intake on the second stage of rat liver
carcinogenesis in a post-initiation bioassay model that
Four-week-old male Wistar rats were obtained from uses preneoplasia detection (GST-P-positive foci) as the
CEMIB (UNICAMP Campinas, SP, Brazil). The animals endpoint [26–29]. The animals were randomly allocated
34 M.C. Dias et al. / Chemico-Biological Interactions 173 (2008) 32–42

Fig. 1. Experimental design of the short-term and medium-term liver bioassays. () 100 or ( ) 200 mg/kg body weight of diethylnitrosamine (DEN);
() 0.9% NaCl; ( ) 70% partial hepatectomy; () diet containing 500 ppm EGb; () diet containing 1000 ppm EGb; n = number of animals/group;
s1–3 : sacrifice 24 h, 2 and 8 weeks after DEN-initiation, respectively.

into five groups. At the beginning of the experiment, 2.3. Biochemical, tissue processing, histology and
groups G1B to G3B were given a single i.p. injection of immunohistochemical procedures
200 mg/kg b.w. of DEN and groups 4B and 5B received
0.9% NaCl. From the 2nd week, groups G2B, G3B and Immediately before necropsy, whole blood was col-
G5B were fed for 6 weeks a diet supplemented with 500, lected and serum enzyme analyses for alanine amino
1000 or 1000 ppm EGb, respectively. Groups G1B and transferase (ALT) and aspartate amino transferase (AST)
G4B were maintained on the basal diet until the end of were carried out spectrophotometrically (Ortho-Clinical
the experiment at week 8. All animals were submitted to Diagnostics, Johnson & Johnson Co., SP, Brazil) to mon-
70% partial hepatectomy (PH) at week 3 and sacrificed itor hepatocellular injury.
at week 8. PH was performed in order to induce vigor- At sacrifice, the liver was removed and weighed
ous regenerative cell replication to increase the initiated after which samples were collected and fixed in
hepatocyte population [29]. 10% phosphate-buffered formalin solution, embedded
In experimental protocols, clinical examinations were in paraffin, cut (5 ␮m thickness) and stained with
performed daily and detailed physical examinations hematoxylin and eosin (H&E). Immunohistochemical
were accomplished weekly. Food and water consump- investigation of glutathione S-transferase P-form (GST-
tions were recorded twice a week (Tuesday/Wednesday P), transforming growth factor alpha (TGF-␣), p53 and
and Thursday/Friday, 18:00 p.m. to 8:00 a.m.) and the proliferating cell nuclear antigen (PCNA) expression
animals were weighed individually once a week through- was performed as follows: Briefly, deparaffinized 5-
out the experimental periods. The diets containing 500 ␮m-thick serial liver sections on poly-l-lysine coated
and 1000 ppm of EGb were prepared weekly, maintained slides were treated with 3% H2 O2 in phosphate-buffered
under −4 ◦ C and offered to the rats daily ad libitum saline for 15 min, nonfat milk for 60 min, polyclonal
to preserve the stability of specific components (mainly anti-rabbit GST-P (Medical and Biological Laboratories
flavonoids) in EGb-supplemented diet. The animals were Co., Tokyo, Japan, clone 311, 1:1000 dilution), mono-
fasted overnight and sacrificed by exsanguination under clonal anti-mouse TGF-␣ (Oncogene Science Inc., New
sodium pentobarbital anesthesia (45 mg/kg p.c.). York, USA, clone Ab-2, 1:200 dilution), monoclonal
M.C. Dias et al. / Chemico-Biological Interactions 173 (2008) 32–42 35

anti-mouse PCNA (Dako Corporation, Carpinterie, CA, apoptotic bodies in liver sections stained with H&E
USA, clone PC10, 1:200 dilution), polyclonal anti-sheep were taken from the literature [30]. For medium-term
p53 (Roche, Mannheim, Germany, clone BGM-1B1, bioassay, the number of clearly discernable TGF-␣-
1:50 dilution) antibodies for 12 h at 4 ◦ C, biotinylated positive FAH per liver unit area was analyzed with
anti-rabbit, anti-mouse or anti-sheep IgG antibodies the aid of KS 300 system as indicated above. PCNA
(Vector Laboratories, Inc., Burlingame, CA, USA, 1:200 labeling and apoptotic indices were determined by eval-
dilution) for 60 min, and streptavidin–biotin–peroxidase uating all S-phase labeled hepatocytes and hepatocytes
solution (TissuGnost Kit was from Merck, Darmstadt, in apoptosis, involving at least 2000 hepatocytes/rat in
Germany, 1:1:50 dilution) for 45 min. Antigen retrieval randomly selected FAH identified by H&E staining. The
was performed for sections submitted to TGF-␣ and p53 immunoreactivity for p53 (wild-type and mutated forms)
immunostaining as follows: for TGF-␣, 0.25% trypsin was also analyzed in the FAH identified by H&E stain-
(Sigma–Aldrich Co., St. Louis, MO, USA) for 15 min ing.
and 0.05% saponin (Calbiochem, La Jolla, CA, USA)
solution for 30 min. For p53, 0.01M citrate buffer (pH 2.5. Statistical analysis
6.0) heated twice, 5 min each time, in a microwave
oven. Chromogen color development was accomplished The statistical analysis was performed using the Jan-
with 3,3 -diaminobenzidine tetrahydrochloride (DAB, del Sigma Stat software (Jandel Corporation, San Rafael,
Sigma-Aldrich Co.) as the substrate to demonstrate CA, USA). Body weight and body-weight gain, absolute
the sites of peroxidase binding. The slides were coun- and relative liver weights, food consumption, number
terstained with Harris’s hematoxylin or H&E. Liver of putative hepatocytes and FAH positive for GST-
sections were processed by omitting incubation with the P and TGF-␣, the mean size and aggregated area of
primary antibodies (GST-P, TGF-␣, p53 and PCNA) as FAH and serum biochemistry data were analyzed by
negative controls for all the immunoreactions. the ANOVA or Kruskal–Wallis tests. PCNA labeling
and apoptosis data were analyzed by the Mann–Whitney
2.4. Immunoreactivity analysis of GST-P, TGF-α, or Kruskal–Wallis tests. Significant differences were
p53 and PCNA markers assumed when P < 0.05.

Single hepatocytes and minifoci (2–15 hepatocytes) 3. Results


positive for GST-P (short-term liver bioassay) and
GST-P-positive FAH larger than 0.15 mm in diame- 3.1. Short-term liver bioassay (anti-initiating
ter (medium-term liver bioassay) were analyzed using screening)
a Nikon photomicroscope (Microphot-FXA) connected
to a KS-300 apparatus (Kontron Elektronic, Germany). No significant alterations in body weight, body-
The liver areas were measured in a special Macro- weight gain or food consumption associated with EGb
Stand device (supported by Canon TV zoom lens ingestion was observed in weeks 1 and 2, before DEN-
V6×16/16–100 mm plus a Canon 58 mm close-up 240 treatment (Table 1). In contrast, body weight and food
lens connected to a CCD black-and-white video camera consumption were significantly reduced in the days
module with a Sony DC-777 camera unit) connected to immediately after DEN initiation (100 mg/kg b.w.) when
the KS-300. Data were expressed as number/cm2 (sin- compared to the non-initiated group (G1A to G3A
gle hepatocytes, minifoci or FAH per liver unit area) vs. G4A, P < 0.001) (data not shown). At the end of
and aggregated area (mm2 /cm2 ) for GST-P-positive week 4, body weight and food consumption of DEN-
FAH. initiated groups had returned to normal levels (data not
For short-term bioassay, immunoreactivity for TGF- shown).
␣ was analyzed in the hepatocytes from zone 3 and/or In animals killed 24 h after DEN administration,
zone 2 of the liver acinus. The PCNA S-phase and p53 all DEN-initiated groups had significantly higher lev-
labeling indices (LI%) were calculated as the percentage els of serum transaminases (AST and ALT) and rates
of labeled hepatocyte nuclei divided by the total num- of cell proliferation, apoptosis and TGF-␣ expression
ber of cells scored (∼2000 hepatocytes). The apoptosis than the non-initiated group (G1A to G3A vs. G4A,
index (AI%) was defined as the number of hepato- P < 0.001) (Table 1, Figs. 2 and 3). Hepatocytes exhibit-
cytes in apoptosis divided by the total number of cells ing immunoreactivity to p53 and TGF-␣ were observed
scored (∼2000 hepatocytes). Criteria for identification in zones 2 and 3 of liver acinus in DEN-initiated
and quantification of hepatocytes in apoptosis and of control group (Fig. 2) while a few centrilobular TGF-
36 M.C. Dias et al. / Chemico-Biological Interactions 173 (2008) 32–42

Table 1
General, liver, biochemical and single/minifoci positive for GST-P data in the short-term bioassay
Parameters Groupsa

DEN 500 EGb + DEN 1000 EGb + DEN Control


(G1A, n = 10) (G2A, n = 10) (G3A, n = 10) (G4A, n = 05)

General datab
Final body weight (g) 255.8 ± 23.91 265.0 ± 30.13 270.00 ± 22.44 280.40 ± 22.44
Body-weight gain (g) 88.60 ± 18.89 97.60 ± 10.74 104.20 ± 12.73 113.60 ± 7.70
Food consumption (g/rat/day) 24.3 ± 2.05 23.70 ± 2.32 23.91 ± 2.18 24.30 ± 2,65
EGb consumption (mg/rat/day) 0 11.85 ± 1.16 23.91 ± 2.18 0
Liver and biochemical datab
Liver absolute weight (g) 10.05 ± 1.36 10.57 ± 1.78 10.79 ± 0.79 12.62 ± 0.93
Relative liver weight (%) 3.93 ± 0.24 3.98 ± 0.39 4.00 ± 0.40 4.50 ± 0.40
AST (U/L) 215.60 ± 43.26* 196.40 ± 31.66* 155.0 ± 38.39*,§ 91.80 ± 20.91
ALT (U/L) 123.80 ± 8.87* 124.60 ± 14.55* 106.4 ± 9.02*,§ 81.60 ± 8.20
GST-P-positive hepatocyte datac
Single (cm2 ) 36.06 ± 6.92 29.16 ± 9.62 18.86 ± 4.04* ND
Minifoci (cm2 ) 4.61 ± 0.53 3.17 ± 0.77 2.38 ± 1.08* ND
Total (cm2 ) 46.84 ± 7.38 41.24 ± 8.36 24.76 ± 5.24* ND

Values are means ± S.D.


* Different from group G4A, 0.05 < P < 0.001.
§ Trend from G1A, 0.05 < P < 0.07; ND: not determined.
a DEN: Diethylnitrosamine (100 mg/kg b.w., i.p.), EGb: Ginkgo biloba extract at 500 and 1000 ppm in basal diet 2 weeks before and 24 h after

DEN-initiation.
b ,c Sacrifice 24 h and 2 weeks after DEN administration, respectively.

␣-positive hepatocytes and no staining for wild-type 3.2. Medium-term liver bioassay (anti-promoting
p53 protein were observed in non-initiated group liver screening)
(data not shown). A reduction of TGF-␣ expression,
which was detected only in zone 3 hepatocytes, was The 6-week post-initiation treatment with EGb did
observed in 1000 ppm EGb DEN-treated animals. This not cause any significant alterations in body weight,
group also presented a significant decrease in the mean body-weight gain or food consumption in both non-
number of PCNA S phase-positive and p53-positive initiated and DEN-initiated groups (Table 2). At the end
hepatocytes and of hepatocytes in apoptosis when com- of the experiment, neither liver weights (absolute and rel-
pared to the liver of the respective control group (G3A ative) nor serum ALT and AST levels were modified by
vs. G1A, 0.05 < P < 0.03 and P = 0.049, respectively) EGb treatment (Table 2). The possible modifying effect
(Figs. 2 and 3). Serum ALT and AST levels were lower of EGb treatment per se on cell proliferation and apop-
in the 1000 ppm EGb DEN-treated group than in the tosis was evaluated in the non-initiated groups (G4B and
respective control group (G3A vs. G1A, P = 0.055 and G5B). Dietary 1000 ppm EGb dose level did not change
0.065, respectively) (Table 1). PCNA labeling or apoptotic indices in the liver (G4B vs.
In the animals killed 2 weeks after DEN-initiation, G5B) (Table 2).
single hepatocytes and minifoci positive for GST-P Post-initiation treatment with EGb did not sup-
were mainly located in zones 2 and 3 and, to a press the development of putative preneoplastic GST-P
lesser extent, in zone 1 of liver acinus in all DEN- or TGF-␣-positive FHA induced by DEN treatment
initiated groups (Fig. 4A and B). At the end of week (Fig. 4C and D, Table 2). PCNA S-phase label-
4, the 1000 ppm EGb treatment, administered before ing and apoptotic indices were not altered into FAH
and during DEN-initiation, had significantly decreased by 6-week EGb treatment. No immunoreactivity for
the mean number of GST-P-positive single hepatocytes p53 was detected in FAH in this early stage of rat
and minifoci per liver unit area (P < 0.001) when com- liver carcinogenesis. Non-initiated groups (G4B and
pared to the respective control group (G3A vs. G1A) G5B) did not develop any GST-P or TGF-␣-positive
(Table 1). FHA.
M.C. Dias et al. / Chemico-Biological Interactions 173 (2008) 32–42 37

Fig. 2. Liver from DEN-initiated group (A, C, E, G) and 1000 ppm EGb plus DEN-initiated group (B, D, F, H), showing apoptosis (A vs. B), PCNA-
positive hepatocytes (C vs. D), p53 nuclear expression (E vs. F) and TGF-␣ expression (G vs. H) in the short-term bioassay. Arrows: Apoptotic
hepatocytes, TV: terminal venule.
38 M.C. Dias et al. / Chemico-Biological Interactions 173 (2008) 32–42

experimental conditions. Importantly, rats fed 1000 ppm


EGb did not present any adverse effects after 6-week
exposure period, as indicated by unaltered body and
liver weights, transaminase levels, liver morphology and
kinetics parameters such as cell proliferation and apop-
tosis rates. This absence of toxicity should be taken into
account if safety measures for public health are to be
implemented in response to increased ingestion of this
herbal drug by human populations, given that EGb has
been clinically prescribed for the treatment of various
diseases [3–6].
The hepatoprotective effect of EGb administra-
tion in the early phase of rat liver carcinogenesis
could be due to a modifying influence on the bio-
transformation/detoxification of DEN, thus reducing
its liver toxicity, mutagenicity, and carcinogenicity.
CYP2E1 is known to be associated primarily with the
biotransformation of a range of compounds, includ-
ing DEN in the liver, but other P450 isozymes
have been found to bioactivate as well [31]. It has
been reported that their biotransformation produces
the promutagenic adducts O6 -ethyldeoxyguanosine, O4
and O6 -ethyldeoxythymidine and 8-hydroxyguanine (8-
OHG) that play a role in the initiation step of rat liver
carcinogenesis [31–34]. Therefore, it could be suggested
that EGb intake before and during initiation with DEN
may provide a protective effect against carcinogenesis,
since this hepatocarcinogen requires metabolic activa-
tion.
After DEN exposure, one hepatic response to
DNA damage and centrilobular cytotoxicity/necrosis is
characterized by regenerative cell proliferation [34].
The response to the damage in both non-initiated
and initiated hepatocytes is associated with the over-
expression of oncogenes/suppressor tumor genes and
growth factors, including p53 and TGF-␣, respectively
Fig. 3. Effects of EGb treatment on liver PCNA labeling (PCNA [35,36].
LI%), p53 labeling (p53 LI%) and apoptotic (AI%) indices in short- Tumor suppressor protein p53 can protect cells from
term bioassay. G1A = DEN, G2A = 500 EGb + DEN, G3A = 1000 growth and division by mediating cell-cycle arrest, DNA
EGb + DEN, G4A = control. DEN: Diethylnitrosamine (100 mg/kg
b.w., i.p.), EGb = Ginkgo biloba extract at 500 and 1000 ppm in
repair and apoptosis [37]. The positivity for p53 in the
basal diet for 2 weeks before and 24 h after DEN-initiation. Val- liver of rats treated with DEN may be attributed to
ues are mean ± S.D., * different from group G4A, 0.05 < P < 0.001; the DNA damage induced by this hepatocarcinogenic
**,*** different from G1A, 0.05 < P < 0.03 and P = 0.049, respectively.
dosage, resulting in cell death and delayed entrance into
critical phases of the cell cycle such as early S or M [35-
4. Discussion 38]. PCNA, a 36kDa molecule that acts as a co-factor for
delta-DNA-polymerase leading to DNA replication, has
The results described herein indicate that EGb intake been considered a feasible marker for liver cell prolifer-
inhibited the initiation but not the promotion stage of ation [39]. Thus, PCNA and p53 analyses are believed to
rat liver carcinogenesis induced by DEN. Therefore, be important indicators of S-phase DNA synthesis and
EGb acted as an anti-initiating agent but did not present cell-cycle progression [37,39]. TGF-␣ is a growth fac-
any suppressing effect on the promotion step of chemi- tor (mitogen stimulus) that activates the protein tyrosine
cally induced rat liver carcinogenesis under the present kinase activity of epidermal growth factor receptor and
M.C. Dias et al. / Chemico-Biological Interactions 173 (2008) 32–42 39

plays an important role in liver regeneration and progres-


sion of carcinogenesis [40,41]. The increase of TGF-␣
expression in DEN-initiated animals could be associ-
ated with the increase in cell proliferation as seen after
tissue deficit created by partial hepatectomy [40]. In the
present study, the beneficial influence of EGb on cell
loss induced by DEN was characterized by a down reg-
ulation of PCNA and TGF-␣ expression, both regulated
by p53 function [39,42]. Therefore, the inhibitory effect
of EGb may be due to its modulation of DEN-induced
DNA damage.
As initiation is a rare event affecting only a few hep-
atocytes, the number of initiated hepatocytes is a major
determinant of the risk for development of liver cancer
and could be used as an endpoint for chemopreven-
tive approaches [26]. Single hepatocytes and minifoci
positive for GST-P develop very early in carcinogen-
treated rats and are mainly located in zones 2 and 3
of liver acinus after a single DEN treatment [24,25].
Although numerical reductions of both single hepato-
cytes and minifoci positive for GST-P were observed in
rats fed 1000 ppm EGb, not all GST-P-positive single
hepatocytes are thought to give rise to putative preneo-
plastic foci and liver tumors [25,43]. Thus, an effective
influence of EGb on the initiation stage of liver carcino-
genesis should continue to be investigated, mainly in
long-term bioassays.
No reports are available on the protective effects of
EGb treatment during the promotion step of chemical
carcinogenesis in rodents. Our data showed that EGb
intake did not modify the development of GST-P and
TGF-␣-positive foci in early phases of rat liver carcino-
genesis. Immunohistochemical expression of TGF-␣ has
been demonstrated in rat liver preneoplastic and neo-
plastic lesions, although a few GST-P-positive FAH
exhibit immunoreactivity for this growth factor [41]. A
selective reduction of TGF-␣-positive foci could rep-
resent a potential chemopreventive property of EGb
since this growth factor may act as a potent hep-
atocyte mitogen during liver carcinogenesis process
[41]. Probably, higher doses of EGb could be more
effective in reducing the clonal expansion of initiated
hepatocytes after DEN initiation or perhaps the anti-
promoting potential of EGb may be expressed in target

Fig. 4. Immunohistochemistry for glutathione S-transferase placen-


tal form (GST-P) (A–C). Putative single hepatocyte (A) and minifoci
(B) positive for GST-P were analyzed in the short-term bioassay. In
addition, foci of altered hepatocytes (FAH) (C) positive for GST-P
were analyzed in the medium-term bioassay. This picture represents
a progressive evolution of the biomarkers during early-phase rat liver
carcinogenesis.
40 M.C. Dias et al. / Chemico-Biological Interactions 173 (2008) 32–42

Table 2
General, liver, biochemical and FAH data in the medium-term bioassay
Parameters Groupsa

DEN DEN + 500 EGb DEN + 1000 EGb Control 1000 EGb
(G1B, n = 10) (G2B, n = 10) (G3B, n = 10) (G4B, n = 05) (G5B, n = 05)

General data
Final body weight (g) 358.98 ± 30.82 357.89 ± 33.12 367.22 ± 29.71 371.88 ± 18.42 380.68 ± 17.00
Body-weight gain (g) 105.06 ± 21.75 102.48 ± 25.29 110.30 ± 22.29 122.10± 14.39 126.68 ± 21.58
Food consumption (g/rat/day) 27.01 ± 2.58 26.70 ± 2.84 27.27 ± 2.52 27.46 ± 3.10 27.20 ± 3.30
EGb consumption (mg/rat/day) 0 13.35 ± 1.42 27.27 ± 2.52 0 27.20 ± 3.30

Liver and biochemical data


Liver absolute weight (g) 9.33 ± 1.33 9.55 ± 1.07 9.37 ± 0.83 9.08 ± 1.09 10.12 ± 0.90
Relative liver weight (%) 2.59 ± 0.22 2.66 ± 0.11 2.55 ± 0.11 2.48 ± 0.25 2.66 ± 0.18
AST (U/L) 85.33 ± 14.6 91.92 ± 22.2 83.58 ± 16.30 82.57 ± 9.54 83.00 ± 7.71
ALT (U/L) 66.75 ± 16.1 69.00 ± 14.1 61.17 ± 8.89 58.43 ± 11.84 55.80 ± 10.18
PCNA LI (%)b 1.75 ± 0.55 1.42 ± 0.65 1.62 ± 0.45 0.42 ± 0.15 0.33 ± 0.05
AI%b 0.15 ± 0.07 0.13 ± 0.05 0.14 ± 0.08 0.08 ± 0.04 0.07 ± 0.03

FHA data
GST-P-positive foci
Number (foci/cm2 ) 9.21 ± 4.44 12.1 ± 4.53 11.65 ± 2.96 0 0
Area (mm2 /cm2 ) 0.23 ± 0.14 0.27 ± 0.15 0.33 ± 0.17 0 0
TGF-␣-positive foci
Number (foci/cm2 ) 1.32 ± 0.35 1.75 ± 0.46 1.65 ± 0.51 0 0

Values are means ± S.D.


a DEN: Diethylnitrosamine (200 mg/kg b.w., i.p.), EGb: Ginkgo biloba extract at 500 and 1000 ppm in basal diet for six weeks; n = number of

rats/group.
b PCNA S-phase-positive hepatocytes and hepatocytes in apoptosis were determined in foci of altered hepatocytes (FHA) (groups G1B to G3B)

and in normal liver (groups G4B and G5B).

organs other than the liver. Given that oxidative stress Acknowledgements
is potentially deleterious to cells and associated with
the progression of many diseases, including cancer This study was supported by CAPES and TOXICAM.
[44], the antioxidant effects derived from crude EGb Dias, M.C. was recipient of a fellowship from CAPES.
or their specific components [7-11] would be a poten-
tial mechanism for cancer prevention in a late phase References
of rat liver carcinogenesis. Since EGb has been shown
to induce cell-cycle arrest and apoptosis in different [1] B.P. Jacobs, W.S. Browner, Ginkgo biloba: a living fossil, Am. J.
human cancer cell lines [15–18], this herbal drug may Med. 108 (2000) 341–342.
be useful for the prevention of liver tumor progres- [2] B. Ahlemeyer, J. Krieglstein, Neuroprotective effects of Ginkgo
biloba extract, Cell Mol. Life Sci. 60 (2003) 1779–1792.
sion. [3] F.V. DeFeudis, V. Papadopoulos, K. Drieu, Ginkgo biloba extracts
In conclusion, the results of the present study and cancer: a research area in its infancy, Fundam. Clin. Pharma-
indicate that EGb presented inhibitory actions during col. 17 (2003) 405–417.
initiation but not post-initiation stage of rat liver car- [4] T. Boonkaew, N.D. Camper, Biological activities of Ginkgo
cinogenesis induced by DEN. Thus, this herbal drug extracts, Phytomedicine 12 (2004) 318–323.
[5] W.R. Zhang, T. Hayashi, H. Kitagawa, Protective effect of ginkgo
presents blocking actions but not suppressing actions extract on rat brain with transient middle cerebral artery occlusion,
during chemically induced liver carcinogenesis [45]. Neurol. Res. 22 (2000) 517–521.
Our findings are interesting for their possible appli- [6] Y. Luo, Ginkgo biloba neuroprotection: therapeutic implications
cation to human cancer chemoprevention among the in Alzheimer’s disease, J. Alzheimers Dis. 3 (2001) 401–407.
population continuously exposed to certain carcino- [7] J. Ding, J. Yu, C. Wang, Ginkgo biloba extract alleviates liver
fibrosis induced by CCl4 in rats, Liver Int. 25 (2005) 1224–1232.
gens. The underlying mechanism(s) of chemoprevention [8] S.X. He, J.Y. Luo, Y.P. Wang, Effects of extract from Ginkgo
of DEN-induced hepatocarcinogenesis still must be biloba on carbon tetrachloride-induced liver injury in rats, World
investigated. J. Gastroenterol. 12 (2006) 3924–3928.
M.C. Dias et al. / Chemico-Biological Interactions 173 (2008) 32–42 41

[9] G. Sener, G.Z. Omurtag, O. Sehirli, Protective effects of Ginkgo [26] M.A. Moore, H. Tsuda, S. Tamano, Marriage of a medium-term
biloba against acetaminophen-induced toxicity in mice, Mol. Cell liver model to surrogate markers: a practical approach for risk and
Biochem. 283 (2006) 39–45. benefit assessment, Toxicol. Pathol. 27 (1999) 237–242.
[10] S.R. Naik, V.S. Panda, Antioxidant and hepatoprotective effects of [27] N. Ito, S. Tamano, T. Shirai, A medium-term rat liver bioassay
Ginkgo biloba phytosomes in carbon tetrachloride-induced liver for rapid in vivo detection of carcinogenic potential of chemicals,
injury in rodents, Liver Int. 27 (2007) 393–399. Cancer Sci. 94 (2003) 3–8.
[11] K. Sasaki, S. Hatta, K. Wada, Effects of extract of Ginkgo biloba [28] F. Pinheiro, R.R. Faria, J.L.V. de Camargo, A.L. Spinardi-
leaves and its constituents on carcinogen-metabolizing enzyme Barbisan, E.F. da Eira, L.F. Barbisan, Chemoprevention of
activities and glutathione levels in mouse liver, Life Sci. 70 (2002) preneoplastic liver foci development by dietary mushroom Agar-
1657–1667. icus blazei Murrill in the rat, Food Chem. Toxicol. 41 (2003)
[12] S.D. Ryu, W.G. Chung, Induction of the procarcinogen-activating 1543–1550.
CYP1A2 by a herbal dietary supplement in rats and humans, Food [29] N. Ito, H. Tsuda, M. Tatematsu, T. Inoue, Y. Tagawa, T. Aoki,
Chem. Toxicol. 41 (2003) 861–866. S. Uwagawa, M. Kagawa, T. Ogiso, T. Masui, Enhancing effect
[13] C. Gaudineau, R. Beckerman, S. Welbourn, K. Auclair, Inhibition of various hepatocarcinogens on induction of preneoplastic glu-
of human P450 enzymes by multiple constituents of the Ginkgo tathione S-transferase placental form positive foci in rats—an
biloba extract, Biochem. Biophys. Res. Commun. 318 (2004) approach for a new medium-term bioassay system, Carcinogen-
1072–1078. esis 9 (1988) 387–394.
[14] T. Sugiyama, Y. Kubota, K. Shinozuka, S. Yamada, K. Yamada, [30] S. Levin, T.J. Bucci, S.M. Cohen, The nomenclature of cell death:
K. Umegaki, Induction and recovery of hepatic drug metabolizing recommendations of an ad hoc Committee of the Society of Tox-
enzymes in rats treated with Ginkgo biloba extract, Food Chem. icologic Pathologists, Toxicol. Pathol. 27 (1999) 484–490.
Toxicol. 42 (2004) 953–957. [31] L. Verna, J. Whysner, G.M. Williams, N-Nitrosodiethylamine
[15] J.C. Chao, C.C. Chu, Effects of Ginkgo biloba extract on cell mechanistic data and risk assessment: bioactivation, DNA-adduct
proliferation and cytotoxicity in human hepatocellular carcinoma formation, mutagenicity, and tumor initiation, Pharmacol. Ther.
cells, World J. Gastroenterol. 10 (2004) 37–41. 71 (1996) 57–81.
[16] Q. Chen, G.W. Yang, L.G. An, Apoptosis of hepatoma cells [32] J.A. Swenberg, D.G. Hoel, P.N. Magee, Mechanistic and
SMMC-7721 induced by Ginkgo biloba seed polysaccharide, statistical insight into the large carcinogenesis bioassays on N-
World J. Gastroenterol. 8 (2002) 832–836. nitrosodiethylamine and N-nitrosodimethylamine, Cancer Res.
[17] B. Ye, M. Aponte, Y. Dai, L. Li, M.C. Ho, A. Vitonis, D. Edwards, 51 (1991) 6409–6414.
T.N. Huang, D.W. Cramer, Ginkgo biloba and ovarian cancer [33] D. Nakae, Y. Kobayashi, H. Akai, N. Andoh, H. Satoh, K. Ohashi,
prevention: epidemiological and biological evidence, Cancer Lett. M. Tsutsumi, Y. Konishi, Involvement of 8-hydroxyguanine
251 (2007) 43–52. formation in the initiation of rat liver carcinogenesis by low
[18] K.S. Kim, K.H. Rhee, J.H. Yoon, J.G. Lee, J.H. Lee, J.B. Yoo, dose levels of N-nitrosodiethylamine, Cancer Res. 57 (1997)
Ginkgo biloba extract (EGb 761) induces apoptosis by the acti- 1281–1287.
vation of caspase-3 in oral cavity cancer cells, Oral Oncol. 41 [34] G.M. Williams, M.J. Iatropoulos, C.X. Wang, Diethylni-
(2005) 383–389. trosamine exposure-responses for DNA damage, centrilobular
[19] A.M. Agha, A.A. El-Fattah, H.H. Al-Zuhair, A.C. Al-Rikabi, cytotoxicity, cell proliferation and carcinogenesis in rat liver
Chemopreventive effect of Ginkgo biloba extract against exhibits some non-linearities, Carcinogenesis 17 (1996) 2253–
benzo(a)pyrene-induced forestomach carcinogenesis in mice: 2258.
amelioration of doxorubicin cardiotoxicity, J. Exp. Clin. Cancer [35] P. Lennartsson, J. Hogberg, U. Stenius, Wild-type p53 expression
Res. 20 (2001) 39–50. in liver tissue and in enzyme-altered foci: an in vivo investiga-
[20] R. Suzuki, H. Kohno, S. Sugie, Preventive effects of extract of tion on diethylnitrosamine-treated rats, Carcinogenesis 19 (1998)
leaves of ginkgo (Ginkgo biloba) and its component bilobalide on 1231–1237.
azoxymethane-induced colonic aberrant crypt foci in rats, Cancer [36] P. Lennartsson, U. Stenius, J. Hogberg, P53 expression and
Lett. 210 (2004) 159–169. TGF-alpha-induced replication of hepatocytes isolated from rats
[21] P. Bannasch, H. Zerban, Predictive value of hepatic preneoplastic exposed to the carcinogen diethylnitrosamine, Cell Biol. Toxicol.
lesions as indicators of carcinogenic response, in: H. Vainio, P. 15 (1999) 31–39.
Magee, D. McGregor, A.J. McMichael (Eds.), Mechanism of Car- [37] A.J. Levine, P53: the cellular gatekeeper for growth and division,
cinogenesis in Risk Identification, IARC Scientific Publication Cell 88 (1997) 323–331.
116, International Agency of Research on Cancer, Lyon, 1992, [38] Y.P. Dragan, J.R. Hully, J. Nakamura, M.J. Mass, J.A. Swenberg,
pp. 389–427. H.C. Pitot, Biochemical events during initiation of rat hepatocar-
[22] Q. Su, P. Bannasch, Relevance of hepatic preneoplasia for human cinogenesis, Carcinogenesis 15 (1994) 1451–1458.
hepatocarcinogenesis, Toxicol. Pathol. 31 (2003) 126–133. [39] J. Foley, T. Ton, R. Maronpot, B. Butterworth, T.L. Goldsworthy,
[23] M. Tatematsu, T. Aoki, M. Kagawa, Y. Mera, N. Ito, Recipro- Comparison of proliferating cell nuclear antigen to tritiated thymi-
cal relationship between development of glutathione S-transferase dine as a marker of proliferating hepatocytes in rats, Environ.
positive liver foci and proliferation of surrounding hepatocytes in Health Perspect. 5 (1993) 199–205.
rats, Carcinogenesis 9 (1988) 221–225. [40] J.E. Mead, N. Fausto, Transforming growth factor alpha may be
[24] M.A. Moore, K. Nakagawa, K. Satoh, T. Ishikawa, K. Sato, Single a physiological regulator of liver regeneration by means of an
GST-P positive liver cells-putative initiated hepatocytes, Carcino- autocrine mechanism, Proc. Natl. Acad. Sci. U.S.A. 86 (1989)
genesis 8 (1987) 483–486. 1558–1562.
[25] K. Satoh, I. Hatayama, N. Tateoka, Transient induction of single [41] M. Gitano, J. Wada, Y. Ariki, M. Kato, H. Wanibuchi, K.
GST-P positive hepatocytes by DEN, Carcinogenesis 10 (1989) Morimura, T. Hidaka, K. Hosoe, S. Fukushima, Possible tumour
2107–2111. development from double positive foci for TGF-alpha and GST-P
42 M.C. Dias et al. / Chemico-Biological Interactions 173 (2008) 32–42

observed in early stages on rat hepatocarcinogenesis, Cancer Sci. [44] E.S. Hwang, G.H. Kim, Biomakers for oxidative stress status of
97 (2006) 478–483. DNA, lipids, and proteins in vitro and in vivo cancer research,
[42] Y. Inoue, T. Tomiya, M. Yanase, P53 may positively regulate Toxicology 229 (2007) 1–10.
hepatocyte proliferation in rats, Hepatology 36 (2002) 336–344. [45] L.W. Wattenberg, Inhibition of tumorigenesis in animals, in:
[43] K. Satoh, I. Hatayama, Anomalous elevation of glutathione B.W. Stewart, D. McGregor, P. Kleihwes (Eds.), Principles of
S-transferase P-form (GST-P) in the elementary process of Chemoprevention, IARC Scientific Publication 139, International
epigenetic initiation of chemical hepatocarcinogenesis in rats, Agency of Research on Cancer, Lyon, 1996, pp. 151–158.
Carcinogenesis 23 (2002) 1193–1198.

Das könnte Ihnen auch gefallen