Você está na página 1de 8

Food and Chemical Toxicology 90 (2016) 10e17

Contents lists available at ScienceDirect

Food and Chemical Toxicology


journal homepage: www.elsevier.com/locate/foodchemtox

Modulating effect of simvastatin on the DNA damage induced by


doxorubicin in somatic cells of Drosophila melanogaster
P.C. Orsolin a, R.G. Silva-Oliveira a, J.C. Nepomuceno a, b, *
a
Universidade Federal de Uberla^ndia, Instituto de Genetica e Bioquímica, Bloco 2E, Campus Umuarama, Uberla ^ndia, Minas Gerais, Brazil
b rio de Patos de Minas, Laborato
Centro Universita rio de Citogenetica e Mutag^
enese, Patos de Minas, Minas Gerais, Brazil

a r t i c l e i n f o a b s t r a c t

Article history: Simvastatin is an antilipemic drug that promotes inhibition of HMG-CoA reductase. Simvastatin can also
Received 3 November 2015 inhibit the formation of other products, such as isoprenoids, conferring additional benefits to this drug,
Received in revised form which include antiproliferative, anti-invasive and pro-apoptotic effects. This study was carried out with
15 January 2016
the aim of evaluating the mutagenic/recombinogenic effect of simvastatin as well as the possible
Accepted 26 January 2016
Available online 29 January 2016
modulatory effects of this statin on the DNA damage induced by doxorubicin (DXR). This analysis was
performed using the somatic mutation and recombination test (SMART) in Drosophila melanogaster. To
study these effects, larvae descendants of both crosses (ST and HB) were chronically treated with five
Keywords:
Simvastatin
concentrations of simvastatin, separately and in association with DXR. The results revealed no muta-
Doxorubicin genic/recombinogenic effect of simvastatin for any of the concentrations tested. A modulating effect of
Drosophila melanogaster simvastatin was also observed on DNA damage induced by DXR. The reduction of total mutant frequency
Antigenotoxicity was observed for spots from descendants of both crosses, but the inhibition was more effective in de-
SMART scendants from the standard cross (ST). It is believed that this modulating effect is mainly associated with
Wing spot assay the antioxidant activity of this class of drugs, although this parameter has not been directly assessed in
this study.
© 2016 Elsevier Ltd. All rights reserved.

1. Introduction mechanisms: a decrease in the endogenous synthesis of cholesterol


and, simultaneously, an increase in receptor synthesis for LDL
Simvastatin is an antilipemic drug of the statin class, which acts cholesterol in hepatic cells, which increases the clearance thereof
to reduce plasma cholesterol levels and, consequently, lowers the (Fonseca, 2005).
risk of atherosclerosis and myocardial infarction (Sparrow et al., There are striking pharmacokinetic differences between the
2001; Tang et al., 2006; Ishikawa et al., 2014). Statins were first statins currently available on the market, including variations in the
derived from the fungi Penicillium citrinum and its mechanism of coefficient of hydrophilicity, hepatic pathway, plasma half-life and
action is inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A efficacy in lowering lipid profiles. Statins also differ regarding in-
reductase (HMG CoA reductase), an enzyme present in the cell's teractions with other drugs that use the same pathway (Fonseca,
endoplasmic reticulum, responsible for the conversion of HMG-CoA 2005; Schachter, 2005). In this context, although all statins have
to mevalonate in the cholesterol biosynthetic pathway (Hindler the same mechanism of action, there are differences in the effec-
et al., 2006; Saito et al., 2008). The main point of limitation and tiveness of responses observed for each statin individually.
control in intracellular cholesterol metabolism occurs precisely at With respect to the chemical structure of statins, there are open
this stage, and is largely influenced by the degree of activity of this or closed lactone ring forms (which makes them structural similar
enzyme (Marie et al., 2008). Inhibition of HMG-CoA reductase to HMG-CoA), constituting the active portion of this class of drugs.
promotes reduction of LDL cholesterol through two principal Simvastatin, specifically, has a closed lactone ring, and is considered
a pro-drug activated in vivo by chemical or enzymatic reaction
(Campo and Carvalho, 2007; Banzato, 2013). In the process of
^ndia, Instituto de Gene
* Corresponding author. Universidade Federal de Uberla - metabolism, the lactone form is converted to the active form hy-
tica e Bioquímica, Bloco 2E, Campus Umuarama, Uberla^ndia, Minas Gerais, Brazil. droxy acid (Chan et al., 2003; Marinho et al., 2012.). Simvastatin is
Tel.: þ55 34 3823 0169; fax: þ 55 34 3821 0300. derived from 2'-methylated lovastatin, a compound isolated from
E-mail address: nepomuceno@ufu.br (J.C. Nepomuceno).

http://dx.doi.org/10.1016/j.fct.2016.01.022
0278-6915/© 2016 Elsevier Ltd. All rights reserved.
P.C. Orsolin et al. / Food and Chemical Toxicology 90 (2016) 10e17 11

the first fungi product, compactin, and is therefore considered a


semisynthetic statin (Levinski and Brown, 2007).
Besides the main action of inhibiting the synthesis of mevalo-
nate, with a consequent reduction in cholesterol levels, simvastatin
can also act by inhibiting the formation of other products, such as
isoprenoids (Demierre et al., 2005), including farnesyl and ger-
anylgeranyl groups, which bind to various proteins (superfamily
Ras/Rho) via prenylation, during the post-translational process
(Edwards and Ericsson, 1999; Yamashita et al., 2008), which gives
the drug additional benefits (pleiotropic). Such benefits include
antiproliferative, anti-invasive, pro-apoptotic and antitumor effects
(Collisson et al., 2002; Liao, 2002; Chan et al., 2003; Sleijfer et al.,
2005; Borahay et al., 2014). According to Ishikawa et al. (2014) Fig. 1. Chemical structure of simvastatin.
the antiproliferative effect of simvastatin is one of the most sig-
nificant, being greater than the effect of other statins such as flu-
other substances was 5% ethanol. All of the dilutions were prepared
vastatin and atorvastatin. The antioxidant action of statins has also
immediately before use. The use of DXR as a positive control in our
been reported in previous studies (Shishehbor et al., 2003;
work, as well as the concentrations used, was based on studies
Mennickent et al., 2008; Gamaleldin et al., 2015).
done previously, which demonstrated generation of reactive oxy-
The extensive knowledge about the genetics of Drosophila mel-
gen species and induction of homologous recombination in D.
anogaster and the long trial experience with this organism have
melanogaster.
made it the primary tool for research on genetic mutation and
toxicology (Sarıkaya and Memmi, 2013). The SMART (Somatic
mutation and recombination test) assay conducted using D. mela- 2.2. Somatic mutation and recombination test (SMART) in
nogaster, is a quick somatic test system, used to detect substances Drosophila melanogaster
that can cause changes in DNA and is also widely used to detect
antimutagenic/antirecombinogenic substances (Graf and Singer, 2.2.1. Stock lineages and crosses
1992; Graf et al., 1998). This bioassay is based on the premise Three mutant lineages of D. melanogaster were used: mwh, flr3
that, during the embryonic development of D. melanogaster, groups and ORR, which possess the genetic markers multiple wing hairs
of cells proliferate mitotically to differentiate into the adult fly body (mwh, 3e0.3) and flare3 (flr3, 3e38.8). More detailed information
structures. If there are genetic changes in the imaginal disc cells, a about the genetic symbols and descriptions of these linkages can be
mutant cell clone will be formed and detected as a smear of mu- found in Lindsley and Zimm (1992).
tants by the wings of the adult fly (Guzman-Rico  n and Graf, 1995). These linkages were submitted to two crossing schemes:
The analysis of these genetic alterations determines the phenotypic
expression of the marker genes mwh or flr3 responsible for changes a) Standard (ST) Cross, in which virgin females flr3/In(3 LR)TM3,
in the trichomes (Graf et al., 1984). ri pp sep I(3)89Aa bx34e and Bds were crossed with males mwh/
Considering the above, the present work was carried out with mwh (Graf et al., 1989);
the main objective of assessing, through the SMART assay, the b) High Bioactivation (HB) Cross, in which virgin females ORR/
mutagenic/recombinogenic effect of simvastatin alone, as well as ORR; flr3/In(3 LR)TM3, ri pp sep I(3)89Aa bx34e and Bds were
the possible modulatory effects of statins on DNA damage induced crossed with males mwh/mwh (Graf and Van Schaik, 1992).
by doxorubicin (DXR). DXR is an antitumor drug of the anthracy-
cline class, which promotes intercalation with the DNA molecule The use of these two types of crosses enables an evaluation of
and inhibition of topoisomerase II (Islaih et al., 2005), and the the mutagenic and anti-mutagenic agents, of direct and indirect
production of free radicals (Nascimento and Martins, 2005; action, according to the basal and elevated levels of the metabo-
Kaiserova  et al., 2006). Antineoplastic DXR is a powerful and lizing enzymes of cytochrome P450. This is possible because the
effective chemotherapeutic agent in the treatment of several types lineage ORR (employed in the HB cross) was created with the aim of
of tumors, but its use is limited due to its cardiotoxicity (Ewer and increasing the performance of the referred to test in the case of
Ewer, 2015). activation of promutagens dependent on activation via cytochrome
P450 (Andrade and Lehmann, 2003).
2. Material and methods From these crosses two types of descendents were obtained:
marked trans-heterozygous (MH, mwh þ/þ flr3), which have wings
2.1. Chemical agents with smooth edges, and balancer heterozygous (BH, mwh þ/þ TM3,
Bds), which have serrated wings (Guzm an-Rinco n and Graf, 1995).
The substance tested was simvastatin (Sinvastacor®), CAS
79902-63-9, batch CV6653, with a molecular formula of C25H38O5 2.2.2. Treatments
and a molecular weight of 418.57 g/mol, produced by Sandoz do The collection of eggs of the descendents of the two crosses
^utica, Cambe
Brasil Indústria Farmace , Parana. Five concentrations described previously, ST and HB, was conducted over a period of
of simvastatin were prepared for use in the experiment: 12.5; 25; 8 h, in vials containing solid agar base and a layer of biological yeast
50; 100 and 200 mM. The chemical structure of the drug tested is supplemented with sucrose. After 72 ± 4 h, third stage larvae were
shown in Fig. 1. washed with reverse osmosis water, collected, and placed in glass
Doxorubicin (Adriblastina®, CAS 25316-40-9, batch 3PL0341, vials containing 1.5 g of instantaneous mashed potato (HIKARI®). To
registered, imported and distributed by the laboratory Pfizer, each tube, 5 mL of simvastatin (in the concentrations 12.5; 25; 50;
Guarulhos, Sa~o Paulo) was used as a positive control. Doxorubicin, 100 and 200 mM) was added, diluted in 5% ethanol, alone and in
with a molecular formula of C27H29NO11-HCl and a molecular association with DXR. The association between DXR and simva-
weight of 580 g/mol, was used at a concentration of 0.125 mg/mL. statin occurred in a co-treatment system.
The negative control used as a reference and for dilution of the These larvae were submitted to a chronic treatment, over a
12 P.C. Orsolin et al. / Food and Chemical Toxicology 90 (2016) 10e17

period of approximately 48 h, during which time they go through substances tested. After this period, the number of flies that
the pupal stage. The larvae were counted (in groups of 100) before hatched in each treatment scheme was counted, and this number
distribution into a series of vials, enabling the toxicity assays to be was then used to calculate the rates of survival after the exposure.
performed. The number of surviving flies per treatment provided In this manner, the survival curves were established based on
an indicator of the toxicity of the compounds. All of the compounds the initial analysis of toxicity, including the percentage of flies that
were tested in duplicate (independent series), conducted at hatch when treated with simvastatin alone and in association with
25  C ± 1  C and relative humidity of approximately 60%. doxorubicin (Fig. 2). The survival percentages of both the controls
were also identified: DXR and 5% ethanol. As can be observed, the
2.2.3. Preparation of the slides and microscopic analysis toxic effect of simvastatin was not identified for any of the con-
After they hatched, the adults emerging from both crosses, centrations analyzed, not even at the highest concentration tested,
possessing trans-heterozygous (mwh þ/þ flr3) and balancer het- since the survival percentage for all of the concentrations was
erozygous (mwh þ/þ TM3, Bds) genotypes, were collected and above 85% (both in the descendents of the ST cross and the HB
preserved in 70% ethanol. The wings were then removed (with the cross). This is why the five concentrations tested - 12.5; 25; 50; 100
aid of entomological forceps) and mounted on coded slides con- and 200 mM - were used for the SMART assay.
taining Faure solution (50 mg of gum arabic, 30 g of chloral hydrate, The results obtained in the test to detect somatic mutation and
30 mL of glycerol, 50 mL of water). These slides were analyzed recombination in D. melanogaster, for the marked trans-
under a light microscope, at a magnification of 400, with the aim heterozygous descendents (MH) and the balancer heterozygous
of identifying mutant spots on different sections of the wings. descendents (BH) of the standard (ST) cross, treated alone and co-
treated with doxorubicin, are presented in Table 1. As expected, the
2.2.4. Statistical analysis positive control (DXR) promoted a statistically significant increase
The Kastenbaum and Bowman (1970) conditional binominal (p < 0.05) in all of the mutant spot categories (single, large, twin
test, at a significance level of 5%, was used to compare the fre- and total number of spots), when compared to the negative control,
quencies of each type of spot (small single, large single or twin) and which confirms its strong activity, above all, in relation to recom-
the total frequency of spots per fly, for each treatment, with this bination, confirmed after analysis of the balancer heterozygous
comparison done in pairs (negative control vs. simvastatin; DXR descendants.
alone vs. simvastatin þ DXR). The multiple-decision procedure In the treatment with simvastatin, none of the five concentra-
described by Frei and Würgler (1988) was used to judge the tions tested (12.5; 25; 50; 100 and 200 mM) showed an increase in
response as positive, weakly positive, negative or inconclusive. All total frequency of spots (p > 0.05), when compared to the negative
of the results with a weak positive diagnosis were analyzed with control, which would suggest the absence of a mutagenic/recom-
the nonparametric U-test by Mann, Whitney and Wilcoxon (Frei binogenic effect for this drug. However, upon evaluating the de-
and Würgler, 1995). The percentage of inhibition of simvastatin scendants treated simultaneously with simvastatin and
was calculated using the frequency of clones per 105 cells, corrected doxorubicin, it was observed that, in all the concentrations tested,
by the control, as follows: [(DXR alone e Simvastatin þ DXR)/DXR simvastatin reduced the frequency of single spots and total number
alone  100] (Abraham, 1994). of spots (when compared to the positive control alone). The per-
Based on the frequency of induction of the mutant spots (per centage of inhibition ranged from 67.07% to 87.55%, and these
105 cells per division), the recombinogenic activity was calculated values were obtained, respectively, for the concentrations of 25 mM
by comparing the pattern of frequency of clones obtained from the and 100 mM. The effect, therefore, was not dose dependent.
MH and BH descendents, as follows: Frequency of mutation Once this modulating effect was identified, the balancer het-
(FM) ¼ frequency of clones in the BH descendents/frequency of erozygous descendants (mwh/TM3) were analyzed, with the aim of
clones in the MH descendents. Frequency of recombination (FR) ¼ 1 identifying the percentages of mutagenic and recombinogenic
e frequency of mutation (FM) (Rezende et al., 2011). events. The comparison between the frequencies of spots of MH
and BH individuals revealed that recombination is the main
3. Results response to the treatment of DXR alone (50.45%). Recombination,
however, was not the main event related to the mutant spots
In this study all compounds were tested in chronic treatments produced by DXR in association with simvastatin (for most of the
(approximately 48 h) on larvae descendants of the ST and HB concentrations). In the concentrations 12.5, 25, 50, 100 and
crosses. All of the compounds were tested in two different exper- 200 mM þ DXR, 40.82%, 53.05%, 34.01%, 31.82% and 32.17% of the
iments, and the results were gathered for the purpose of statistical spots were due to recombinogenetic changes, respectively. There-
analyses, after checking that there was no heterogeneity in the fore, these data indicate that simvastatin, in the presence of basal
samples. levels of cytochrome P450, significantly reduces the number of
Before the analysis using the SMART assay, simvastatin was mutant spots, altering (reducing) the recombinogenetic profile of
submitted to a toxicity test in D. melanogaster, with the aim of doxorubicin.
evaluating the percentage of survival of the individuals treated The results for the descendents (MH and BH) of the high bio-
with the different concentrations used. The toxicity is determined activation (HB) cross, treated alone and in association with doxo-
based on the number of larvae that do not reach the adult phase. rubicin are presented in Table 2. Once again, a significant increase
This analysis is crucial since the reduction in the percentage of (p < 0.05) was found in the total frequency of spots identified in the
larval survival is an indication that the compounds affected the positive control, when compared to the frequency of the negative
development of the larvae and also that the number of adults control. DXR, therefore, induced the appearance of mutant spots,
emerging should be high enough to allow for the subsequent presenting primarily recombinogenic activity (69.92%).
treatments (Demir et al., 2013). In the treatment with simvastatin, the results revealed no dif-
Concentrations used in this test were initially delineated based ference between the total frequency of mutant spots and the fre-
on a study carried out by Spindler et al. (2012), using D. mela- quency identified in the negative control (p > 0.05) for the five
nogaster. Once the concentrations are established, the larvae, de- concentrations tested. These results suggest, therefore, that, even
scendents of the standard and high bioactivation crosses, were after intense metabolic biotransformation, simvastatin does not
submitted to a chronic treatment (for approximately 48 h) with the possess mutagenic and/or recombinogenic effect in D.
P.C. Orsolin et al. / Food and Chemical Toxicology 90 (2016) 10e17 13

Simvastatin
100

90

Survival (%)
80
ST
HB
70

60

50
5% Ethanol 12,5 μM 25 μM 50 μM 100 μM 200 μM

Concentrations

Simvastatin + DXR
100

90
Survival (%)

80
ST
70
HB
60

50
DXR (0,125 12,5 μM 25 μM 50 μM 100 μM 200 μM
mg/mL) +DXR +DXR +DXR +DXR +DXR

Concentrations
Fig. 2. Percentage of survival of flies after metamorphosis of larvae 72 h treated with different concentrations of simvastatin, alone and in combination with DXR.

melanogaster. demonstrated here corroborate previous results, also identified


While the analysis of descendants co-treated with simvastatin using the SMART assay in D. melanogaster, performed with two
and doxorubicin show that in the highest concentration tested, synthetic statins (atorvastatin and rosuvastatin), in our laboratory,
200 mM, simvastatin significantly reduced (p < 0.05) the total fre- and reported in a previous publication (Orsolin et al., 2015).
quency of spots in comparison with the frequency observed in the In the research cited above, the results demonstrate the absence
individuals treated only with DXR (positive control). In the other of a mutagenic effect for atorvastatin and rosuvastatin and a
concentrations no statistically significant reductions were observed modulating effect on the damage to DNA induced by DXR. This
in the total frequency of spots. effect was found in all the concentrations tested in the descendents
The results also reveal that simvastatin, at a concentration of of the ST and HB crosses treated with rosuvastatin, and only in
200 mM, in the descendants of the HB cross, reduced the frequency descendents of the HB cross treated with atorvastatin.
of spots by 65.48%. With regard to the proportion of spots obtained In a study carried out by Peron et al. (2008), with the aim of
by mutation and recombination, it was observed that 64.52% of the evaluating the cytotoxicity and mutagenicity of simvastatin in the
mutant spots are related to recombinogenetic alterations. There- bone marrow cells of Wistar rats, treated in vivo, and in the meri-
fore, these data suggest that simvastatin, although it significantly stematic cells of the root of Allium cepa L., it was also found that
reduced the number of mutant spots at the highest concentration simvastatin does not present cytotoxic action, nor mutagenic effect,
tested, did little to alter the recombinogenetic profile of DXR, after since it did not increase the number of chromosome aberrations.
intense metabolic biotransformation. Under the present experimental conditions it was observed that
simvastatin, in addition to not presenting a mutagenic/recombi-
nogenic effect, presented a modulating response to the activity of
4. Discussion doxorubicin. This response was observed in the descendants of
both standard and high bioactivation crosses. Although inhibition
The present study investigated the mutagenic and recombino- was more effective in the descendents of the ST cross. The differ-
genic action of simvastatin, alone, as well as the possible modu- ences between the two crosses, ST and HB, are related to the levels
lating effects of this statin on damage to DNA induced by of cytochrome P450.
doxorubicin in the somatic cells of D. melanogaster. The effects
14 P.C. Orsolin et al. / Food and Chemical Toxicology 90 (2016) 10e17

Table 1
Summary of results obtained in the marked trans-heterozygous descendants (MH) and balancer-heterozygous (BH) of Drosophila melanogaster derived from the standard (ST)
cross treated with different simvastatin concentrations (separately and in combination with DXR), positive control (DXR 0.125 mg/mL) and negative control (5% ethanol).

Treatments Number Spots per fly (N0 of spots); statistical diagnosisa Spots with Frequency of clone formation/ Recombination
of flies mwh clonec 105 cells per cell divisiond (%)
(N) (n)
DXR Simvastatin Small single Large single Twin Total Observed Control Inhibitione
(mg/mL) (mM) ls)b
(1e2 ce (>2 cels)b m¼5 spots corrected (%)
m¼2 m¼5 m¼2

mwh/flr3

0 0 60 0.38 (23) 0.02 (1) 0.00 (0) 0.40 (24) 24 0.82

0 12.5 60 0.33 (20)- 0.00 (0)i 0.03 (2)i 0.37 (22)- 22 0.75 0.07
0 25 60 0.32 (19)- 0.03 (2)i 0.00 (0)i 0.35 (21)- 21 0.72 0.10
0 50 60 0.38 (23)- 0.07 (4)i 0.00 (0)i 0.45 (27)- 27 0.92 0.10
0 100 60 0.40 (24)- 0.02 (1)i 0.00 (0)i 0.42 (25)- 25 0.85 0.03
0 200 60 0.47 (28)i 0.00 (0)i 0.02 (1)i 0.48 (29)- 27 0.92 0.10
0.125 0 60 1.65 (99)þ 0.20 (12)þ 0.10 (6)þ 1.95 (117)þ 97 3.31 2.49 50.45
0.125 12.5 60 0.43 (26)* 0.22 (13)i 0.08 (5)i 0.73 (44)* 43 1.47 0.65 40.82 73.90
0.125 25 60 0.53 (32)* 0.13 (8)i 0.13 (8)i 0.80 (48)* 48 1.64 0.82 53.05 67.07
0.125 50 60 0.42 (25)* 0.22 (13)i 0.10 (6)i 0.73 (44)* 43 1.47 0.65 34.01 73.90
0.125 100 60 0.33 (20)* 0.17 (10)i 0.07 (4)i 0.57 (34)* 33 1.13 0.31 31.86 87.55
0.125 200 60 0.37 (22)* 0.18 (11)i 0.15 (9)i 0.70 (42)* 42 1.43 0.61 32.17 75.50
mwh/TM3
f
0 0 40 0.28 (11) 0.00 (0) 0.28 (11) 11 0.56
0.125 0 40 0.68 (27)þ 0.13 (5)þ 0.80 (32)þ 32 1.64 1.08
0.125 12.5 40 0.43 (17)i 0.00 (0)* 0.43 (17)* 17 0.87 0.31
0.125 25 40 0.38 (15)* 0.00 (0)* 0.38 (15)* 15 0.77 0.20
0.125 50 40 0.45 (18)i 0.03 (1)i 0.48 (19)* 19 0.97 0.41
0.125 100 40 0.38 (15)* 0.00 (0)* 0.38 (15)* 15 0.77 0.20
0.125 200 40 0.48 (19)i 0.00 (0)* 0.48 (19)* 19 0.97 0.41

Marker-trans-heterozygous flies (mwh/flr3) and balancer-heterozygous flies (mwh/TM3) were evaluated.


*p  0.05 vs. DXR only.
a
Statistical diagnoses according to Frei and Würgler (1995). U-test, two sided; probability levels: -, negative; þ, positive; i, inconclusive; p  0.05 vs. negative control (5%
ethanol).
b
Including rare single flr3 spots.
c
Considering the mwh clones for the single spots and mwh for the twin spots.
d
Frequency of clone formation: clones/flies/48,800 cells (without size correction).
e
Calculated as [(DXR alone e Statin þ DXR/DXR alone)  100], according to Abraham (1994).
f
Balancer chromosome TM3 does not carry the flr3 mutation and recombination is suppressed, due to the multiple inverted regions in these chromosomes.

The HB cross used the lineage ORR, which presents chromo- generate active metabolites with a mutagenic/recombinogenic ef-
somes 1 and 2 from the lineage Oregon R, which is resistant to DDT, fect, the product resulting from the metabolic biotransformations
and exhibits high expression of the cytochrome P450 enzymes of simvastatin, when associated with doxorubicin (in a co-
€lich and Wrügler, 1989; Graf and Van Schaik, 1992). Whereas
(Fro treatment system), reduced the total number of mutant spots.
the descendants of the ST cross have basal levels of cytochrome However, with less effective inhibition (observed only at the
P450. According to Andrade and Lehmann (2003), the lineage ORR highest concentration) to that obtained without metabolic trans-
was developed with the aim of increasing the performance of the formation (observed at all of the concentrations tested).
SMART assay for the activation of promutagens that depend on The mechanisms by which simvastatin reduces the damage to
activation via cytochrome P450. DNA induced by DXR were not directly evaluated in this study.
Simvastatin is a prodrug, administered in the form of an active However, the use of the co-treatment system enables an analysis of
lactone, requiring hepatic transformation for conversion into b- the effect of simvastatin on stages that occur before the harm
hydroxy acid, its active metabolite (Sabina et al., 2012; Gazzerro induced by DXR, enabling an evaluation of its modulating effect. As
et al., 2012). Most of the statins, including simvastatin, are metab- such, it can be assumed, based on the results obtained in the pre-
olized by enzymes from the cytochrome P450 system (CYP3A4) sent study, that the active metabolites of simvastatin, resulting
(Gazzerro et al., 2012). CYP34A is the most abundant isoenzyme of from the process of biotransformation, are capable of interacting
cytochrome P450 in the liver and small intestine; it acts on the directly with the active groups of the DXR, altering the inhibition
oxidative metabolism of around 50% of all the drugs available on response, as seen in the descendants of the standard (ST) cross. This
the market (Granvil et al., 2003). The CYP3A4 of mammals is is a possibility, though one that cannot be explained precisely in the
analogous to the family CYP6 in Drosophila (Tijet et al., 2001). present study, since it is not possible to identify how this interac-
Through these systems of biotransformation, like cytochrome tion between the two substances occurs.
P450, biologically inactive substances can be introduced into the The results for the individuals treated with DXR confirmed the
body and converted into active metabolites due to alterations in recombinogenic effect of this drug. With regard to the action
their structure. Although the purpose of deep biotransformation of mechanism, doxorubicin binds strongly to DNA due to its ability to
the xenobiotics is detoxification, the metabolite is not always less intercalate between base pairs. When intercalating in a DNA
toxic than the compound itself (Guecheva and Henriques, 2003). molecule, DXR promotes breakage in the molecule, inhibiting the
The process of biotransformation can generate toxic intermediaries, synthesis of DNA and RNA (Craig and Stitzel, 2005; Kaiserova  et al.,
electrophilics, mutagenics or carcinogenics (Hodgson and Rose, 2006), while also promoting the production of free radicals
2007). (Nascimento and Martins, 2005). According to Lehmann et al.
In the present study, although biotransformation did not (2003), the intrinsic link between inhibition of topoisomerases,
P.C. Orsolin et al. / Food and Chemical Toxicology 90 (2016) 10e17 15

Table 2
Summary of results obtained in the marked trans-heterozygous descendants (MH) and balancer-heterozygous (BH) of Drosophila melanogaster derived from the high bio-
activation (HB) cross treated with different simvastatin concentrations (separately and in combination with DXR), positive control (DXR 0.125 mg/mL) and negative control (5%
ethanol).

Treatments Number Spots per fly (N0 of spots); statistical diagnosisa Spots with Frequency of clone formation/ Recombination
of flies mwh clonec 105 cells per cell divisiond (%)
(N) (n)
DXR Simvastatin Small single Large single Twin Total Observed Control Inhibitione
(mg/mL) (mM) ls)b
(1e2 ce ls)b
(>2 ce m¼5 spots m ¼ 2 corrected (%)
m¼2 m¼5

mwh/flr3

0 0 60 1.10 (66) 0.03 (02) 0.02 (1) 1.15 (69) 69 2.36

0 12.5 60 1.12 (67)- 0.08 (5)i 0.02 (1)i 1.22 (73)- 71 2.42 0.07
0 25 60 1.05 (63)- 0.08 (5)i 0.03 (2)i 1.17 (70)- 70 2.39 0.03
0 50 60 1.30 (78)- 0.08 (5)i 0.00 (0)i 1.38 (83)- 82 2.80 0.44
0 100 60 1.20 (72)- 0.07 (4)i 0.05 (3)i 1.32 (79)- 79 2.70 0.34
0 200 60 1.32 (79)- 0.03 (2)i 0.02 (1)i 1.37 (82)- 82 2.80 0.44
0.125 0 60 1.48 (89)- 2.20 (132)þ 0.10 (6)i 3.78 (227)þ 225 7.68 5.33 69.92
0.125 12.5 60 1.43 (86) ns 1.38 (83)* 0.13 (8)i 2.95 (177) ns 173 5.91 3.55
0.125 25 60 1.48 (89) ns 1.35 (81)* 0.07 (4)i 2.90 (174) ns 174 5.94 3.59
0.125 50 60 1.27 (76) ns 1.55 (93)* 0.25 (15)* 3.07 (184) ns 182 6.22 3.86
0.125 100 60 1.40 (84) ns 1.05 (63)* 0.25 (15)* 2.70 (162) ns 161 5.50 3.14
0.125 200 60 1.22 (73) ns 0.77 (46)* 0.08 (5)i 2.07 (124)* 123 4.20 1.84 64.52 65.48
mwh/TM3
f
0 0 40 0.38 (15) 0.00 (0) 0.38 (15) 15 0.77
0.125 0 40 0.98 (39)i 0.15 (6)i 1.13 (45)þ 45 2.31 1.54
0.125 200 40 0.73 (29)i 0.00 (0)* 0.73 (29)* 29 1.49 0.72

Marker-trans-heterozygous flies (mwh/flr3) and balancer-heterozygous flies (mwh/TM3) were evaluated.


*p  0.05 vs. DXR only, ns: not significant.
a
Statistical diagnoses according to Frei and Würgler (1995). U-test, two sided; probability levels: -, negative; þ, positive; i, inconclusive; p  0.05 vs. negative control (5%
ethanol).
b
Including rare single flr3 spots.
c
Considering the mwh clones for the single spots and mwh for the twin spots.
d
Frequency of clone formation: clones/flies/48,800 cells (without size correction).
e
Calculated as [(DXR alone e Statin þ DXR/DXR alone)  100], according to Abraham (1994).
f
Balancer chromosome TM3 does not carry the flr3 mutation and recombination is suppressed, due to the multiple inverted regions in these chromosomes.

blocking of the synthesis and replication of DNA and inducing by DXR, this reduction may also be associated with the apoptotic
double breaks, associated with the fact that this latter event is one effect of this drug. In a study conducted by Yanae et al. (2011), it was
of the main substrates for the occurrence of recombination, also demonstrated that statins inhibit cellular proliferation and induce
make DXR a potent recombinogenic agent. This recombinogenic apoptosis in the glioma cells of rats. An increase in the activity of
effect has already been reported in a series of previous studies caspase-3, an enzyme with a central role in this process, was also
performed on the somatic cells of D. melanogaster (Lehmann et al., observed. In addition, the statins reduced the levels of Ras regu-
2003; Costa and Nepomuceno, 2006; Rezende et al., 2009; Orsolin lated by the ERK1/2 and Akt pathways. According to the authors
et al., 2012, 2015). cited, these findings indicate that statins can be used as anti-
Another possible mechanism to explain the reduction in the carcinogenic agents in glioblastoma.
number of mutant spots produced by simvastatin, when associated Takeda et al. (2007) also demonstrated that simvastatin is
with DXR, could be associated with the antioxidant effect of statins. capable of suppressing cellular survival and the invasive activity of
Elbaky et al. (2006) investigated the possible protective effects of squamous carcinoma cells of the head and neck by activating
simvastatin on the nephrotoxicity induced by adriamycin in rats integrin b1 (involved in the ERK signaling cascade). These authors
and found that the administration of simvastatin, before and con- also confirmed the activation of the inhibitors of CDK, p21 and p27,
current with adriamycin, promoted a reduction in lipid peroxida- and the inhibitors of caspase-3. According to Bardou et al. (2010),
tion and an increase in the activity of antioxidant enzymes. statins, in the case of colorectal cancer, may counter the growth of
Similarly, Elbaky et al. (2010) investigated possible protective cancer by inducing apoptosis, by reducing the expression of anti-
effects of simvastatin on the cardiotoxicity induced by doxorubicin. apoptotic proteins like BC12 or clAP1 and by increasing the
These authors found that the administration of simvastatin also expression of pro-apoptotic proteins, like BMP, for example.
significantly reduced the myocardial alterations induced by treat- As a result of the effects described above, statins, including
ment with DXR. These results suggest that simvastatin offers pro- simvastatin, have been highlighted for their possible antitumor
tection against cardiac damage induced by DXR, in terms of effects. In this sense, Bardou et al. (2010) affirmed that there are
oxidative stress. various mechanisms that could justify the antitumor effects of
Ungureanu et al. (2003) observed that individuals with hyper- statins, including the induction of apoptosis (through different
cholesterolemia, who received daily doses of simvastatin (10 mg/ pathways), inhibition of cellular growth, in addition to angiogen-
day, for 8 months), in addition to a significant reduction in total esis and an improvement of the immune response.
cholesterol, presented a significant improvement in some anti- In conclusion, the results reported showed that, under the
oxidant enzyme parameters: superoxide dismutase, glutathione present experimental conditions, simvastatin presented a modu-
peroxidase and catalase. lating effect on the damage to DNA induced by doxorubicin in the
In addition to the recognized antioxidant effect of simvastatin, somatic cells of D. melanogaster. It is believed that this modulating
which may have resulted in the reduction of mutant spots induced effect is associated with the apoptotic and, primarily, antioxidant
16 P.C. Orsolin et al. / Food and Chemical Toxicology 90 (2016) 10e17

activities of this class of drugs, although these effects have not been Graf, U., Würgler, F.E., Katz, A.J., Frei, H., Juon, H., Hall, C.B., Kale, P.G., 1984. Somatic
mutation and recombination test in Drosophila melanogaster. Environ. Mutagen
directly evaluated in this study. It is suggested, therefore, that new
6, 153e188.
studies involving other test organisms and experimental models be Granvil, C.P., Yu, A.M., Elizondo, G., Akiyama, T.E., Cheung, C., Feigenbaum, L.,
conducted with the aim of promoting a better understanding of the Kraus, K.W., Gonzales, F.J., 2003. Expression of the human CYP3A4 gene in the
modulating activity of simvastatin in relation to substances that small intestine of transgenic mice: in vitro metabolism and pharmacokinetics of
midazolam. Cancer Lett. 167, 99e104.
induce damage to DNA, such as doxorubicin. Guecheva, T.K., Henriques, J.A.P., 2003. Metabolismo de xenobio  ticos: citocromo P-
450. In: Silva, J., Erdtmann, B., Henriques, J.A.P. (Eds.), Gene tica Toxicolo gica.
Alcance, Porto Alegre, pp. 223e247. Cap.11.
Transparency document Guzma n-Ricon, J., Graf, U., 1995. Biomonitors and Biomarkers as Indicators of
Environmental Change. Phenunm Press, New York, pp. 169e181.
Hindler, K., Eltzschig, H.K., Fox, A.A., Body, S.C., Shernan, S.K., Collard, C.D., 2006.
Transparency document related to this article can be found
Influence of statins on perioperative outcomes. J. Cardiothorac. Vasc. Anesth. 20,
online at http://dx.doi.org/10.1016/j.fct.2016.01.022. 251e258.
Hodgson, E., Rose, R.L., 2007. The importance of cytochrome P450 2B6 in the human
metabolism of environmental chemicals. Pharmacol. Ther. 113, 420e428.
References Ishikawa, S., Hayashi, H., Kinoshita, K., Abe, M., Kuroki, H., Tokunaga, R.,
Tomiyasu, S., Tanaka, H., Sugita, H., Arita, T., Yagi, Y., Watanabe, M., Hirota, M.,
Abraham, S.K., 1994. Antigenotoxicity of coffee in the Drosophila assay for somatic Baba, H., 2014. Statins inhibit tumor progression via an enhancer of zeste ho-
mutation and recombination. Mutagenesis 9, 383e386. molog 2-mediated epigenetic alteration in colorectal cancer. Int. J. Cancer 135,
Andrade, H.H.R., Lehmann, M., 2003. Teste para detecça ~o de mutaça ~o e 2528e2536.
recombinaç~ ao som atica em Drosophila melanogaster. In: Ribeiro, L.R., Islaih, M., Halstead, B.W., Kadura, I.A., Li, B., Reid-Hubbard, J.L., Flick, L., Altizer, J.L.,
Salvadori, D.M.F., Marques, E.K. (Eds.), Mutage ^nese Ambiental. Ulbra, Canoas, Thom Deahl, J., Monteith, D.K., Newton, R.K., Watson, D.E., 2005. Relationships
pp. 281e307. between genomic, cell cycle, and mutagenic responses of TK6 cells exposed to
Banzato, T.P., 2013. Avaliaça ~o da toxicidade reprodutiva da sinvastatina em ratos DNA damaging chemicals. Mutat. Res. 578, 100e116.
adultos. Dissertaça ~o (Biologia Geral e Aplicada). Instituto de Biocie ^ncias, Uni- Kaiserova , H., Den Hartog, G.J.M., Simunek, T., Schroterov a, L., Kvasnickov a, E.,
versidade Estadual Paulista, Botucatu. Bast, A., 2006. Iron is not involved in oxidative stress-mediated cytotoxicity of
Bardou, M., Barkun, A., Martel, M., 2010. Effect of statin therapy on colorectal cancer. doxorubicin and bleomycin. Br. J. Pharmacol. 149, 920e930.
Gut 59, 1572e1585. Kastenbaum, M.A., Bowman, K.O., 1970. Tables for determining the statistical sig-
Borahay, M.A., Kilic, G.S., Yallampalli, C., Snyder, R.R., Hankins, G.D.V., Al-Hendy, A., nificance of mutation frequencies. Mutat. Res. 9, 527e549.
Boehning, D., 2014. Simvastatin potently induces calcium-dependent apoptosis Lehmann, M., Franco, A., Vilar, K.S.P., Reguly, M.L., Andrade, H.H.R., 2003. Doxoru-
of human leiomyoma cells. J. Biol. Chem. 51, 35075e35086. bicin and two of its analogues are preferential inducers of homologous
Campo, V.L., Carvalho, I., 2007. Estatinas hipolipe ^micas e novas tende ^ncias ter- recombination compared with mutational events in somatic cells of Drosophila
ape^uticas. Quím. Nova 30, 01e14. melanogaster. Mutat. Res. 539, 167e175.
Chan, K.K., Oza, A.M., Siu, L.L., 2003. The statins as anticancer agents. Clin. Cancer Levinski, R.J., Brown, H., 2007. Statins. Too many people are taking them (and
Res. 9, 10e19. they're doing far less good than you think). Lancet 69, 268e269.
Collisson, E.A., Carranza, D.C., Chen, I.Y., Kolodney, M.S., 2002. Isoprenylation is Liao, J.K., 2002. Beyond lipid lowering: the role of statins in vascular protection. Int.
necessary for the full invasive potential of RhoA overexpression in human J. Cardiol. 86, 05e18.
melanoma cells. J. Investigative Dermatol. 119, 1172e1176. Lindsley, D.L., Zimm, G.G., 1992. The Genome of Drosophila melanogaster, first ed.
Costa, W.F., Nepomuceno, J.C., 2006. Protective effects of a mixture of antioxidant Academic Press, San Diego. 1133pp.
vitamins and mineral on the genotoxicity of doxorubicin in somatic cells of Marie, I., Delafenetre, H., Massy, N., Thuillez, C., Noblet, C., 2008. Tendinous disor-
Drosophila melanogaster. Environ. Mol. Mutagen 47, 18e24. ders attributed to statins: a study on ninety-six spontaneous reports in the
Craig, C.R., Stitzel, R.E., 2005. Farmacologia Moderna: Com Aplicaço ~es Clínicas, 6. ed. period 1990-2005 and review of the literature. Arthritis Rheum. 59, 367e372.
Guanabara Koogan, Rio de Janeiro. 815 pp. Marinho, F.D.M., Diniz, M.M.L., Zanon, J. C. da C., Reis, I.A., Lima, A.A., Soares, C.D.V.,
Demierre, M.F., Higgins, P.D., Gruber, S.B., Hawk, E., Lippman, S.M., 2005. Statins and 2012. Influe ^ncia da temperatura em estudos de dissoluça ~o de formas farm-
cancer prevention. Nat. Rev. Cancer 5, 930e942. ace^uticas contendo sinvastatina. Rev. Bras. Farm. 93, 38e42.
Demir, E., Turna, F., Kaya, B., Creus, A., Marcos, R., 2013. Mutagenic/recombinogenic Mennickent, S.C., Bravo, M.D., Calvo, C.M., Avello, M.L., 2008. Efectos pleiotro picos
effects of four lipid peroxidation products in Drosophila. Food Chem. Toxicol. de las estatinas. Rev. Med. Chile 136, 775e782.
53, 221e227. Nascimento, M.C.M.O., Martins, A.S., 2005. Cardiomiopatia induzida pela adriami-
Edwards, P.A., Ericsson, J., 1999. Sterols and isoprenoids: signaling molecules cina: uma revis~ ao. Arq. Cie^ncias Saúde 12, 111e115.
derived from the cholesterol biosynthetic pathway. Annu. Rev. Biochem. 68, Orsolin, P.C., Silva-Oliveira, R.G., Nepomuceno, J.C., 2012. Assessment of the muta-
157e185. genic, recombinagenic and carcinogenic potential of orlistat in somatic cells of
Elbaky, N.A.A., Hammad, L.N., Atia, T.A., 2006. Protective effect of simvastatin Drosophila melanogaster. Food Chem. Toxicol. 50, 2598e2604.
against adriamycin-Induced nephrotoxicity in rats; biochemical and histologi- Orsolin, P.C., Silva-Oliveira, R.G., Nepomuceno, J.C., 2015. Modulating effect of syn-
cal study. Egypt. J. Hosp. Med. 25, 725e739. thetic statins against damage induced by doxorubicin in somatic cells of
Elbaky, N.A.A., Ali, A.A., Ahmed, R.A., 2010. Cardioprotective effect of simvastatin on Drosophila melanogaster. Food Chem. Toxicol. 81, 111e119.
doxorubicin -induced cardiotoxicity in rats. J. Basic Appl. Sci. 6, 1e11. Peron, A.P., Felipes, J., Oliveira, A.C.S., Vicentin, V.E.P., 2008. Investigaç~ ao da cit-
Ewer, M.S., Ewer, S.M., 2015. Cardiotoxicity of anticancer treatments. Nat. Rev. otoxicidade e mutagenicidade do hipocolesteremiante sinvastatina em ce lulas
Cardiol. 12, 547e558. de ratos wistar e de Allium cepa L. Rev. Bras. Toxicol. 21, 93e96.
Fonseca, F.A.H., 2005. Farmacocine tica das estatinas. Arq. Bras. Cardiol. 85, 09e14. Rezende, A. A. de, Graf, U., Guterres, Z.R., Kerr, W.E., Spano  , M.A., 2009. Protective
Frei, H., Würgler, F.E., 1988. Statistical methods to decide whether mutagenicity test effects of proanthocyanidins of grape (Vitisvinifera L.) seeds on DNA damage
data from Drosophila assay indicate a positive, negative or inconclusive result. induced by Doxorubicin in somatic cells of Drosophila melanogaster. Food Chem.
Mutat. Res. 203, 297e308. Toxicol. 47, 1466e1472.
Frei, H., Würgler, F.E., 1995. Optimal experimental design and sample size for the Rezende, A. A. de, Silva, M.L.A., Tavares, D.C., Cunha, W.R., Rezende, K.C.S.,
statistical evaluation of data from somatic mutation and recombination test Bastos, J.K., Lehmann, M., Andrade, H. H. R. de, Guterres, Z.R., Silva, L.P.,
(SMART) in Drosophila. Mutat. Res. 334, 247e258. Spano , M.A., 2011. The effect of the dibenzylbutyrolactoliclignan (-)-cubebin on
€lich, A., Würgler, F.E., 1989. New tester strains with improved bioactivation ca-
Fro doxorubicin mutagenicity and recombinogenicity in wing somatic cells of
pacity for the Drosophila wing-spot test. Mutat. Res. 216, 179e187. Drosophila melanogaster. Food Chem. Toxicol. 49, 1235e1241.
Gamaleldin, I.H., Fars, K.A., Sabry, M.A., Gamal, A.O., 2015. Influence of simvastatin Sabina, A.G., Da vila, J.G., Serrano, P.S., Juanetey, C.G., Pacheco, R.M., 2012. Consid-
chronotherapy on erythrocytes nitric oxide synthase activity. Int. J. Pharmacol. eraciones específicas en la prescripcio n e intercambio terape utico de estatinas.
5, 448e455. Farm. Hosp. 36, 97e108.
Gazzerro, P., Proto, M.C., Gangemi, G., Malfitano, A.M., Ciaglia, E., Pisanti, S., Saito, A., Saito, N., Mol, W., Furukawa, H., Tsutsumida, A., Oyama, A., Sekido, M.,
Santoro, A., Laezza, C., Bifulco, M., 2012. Pharmacological actions of statins: a Sasaki, S., Yamamoto, Y., 2008. Simvastatin inhibits growth via apoptosis and
critical appraisal in the management of cancer. Pharmacol. Rev. 64, 102e146. the induction of cell cycle arrest in human melanoma cells. Melanoma Res. 18,
Graf, U., Abraham, S.K., Guzma n-Rinco  n, J., Würgler, F.E., 1998. Antigenotoxicity 85e94.
studies in Drosophila melanogaster. Mutat. Res. 402, 203e209. Sarıkaya, R., Memmi, B.K., 2013. Detection of transfluthrin and metofluthrin geno-
Graf, U., Frei, H., K€agi, A., Katz, A.J., Würgler, F.E., 1989. Thirty compounds tested in toxicity in the ST cross of the Drosophila Wing Spot Test. Chemosphere 93,
the Drosophila wing spot test. Mutat. Res. 222, 359e373. 238e242.
Graf, U., Singer, D., 1992. Genotoxicity testing of promutagens in the wing somatic Schachter, M., 2005. Chemical, pharmacokinetic and pharmacodynamic properties
mutation and recombination test in Drosophila melanogaster. Rev. Int. Contam. of statins: an update. Fundam. Clin. Phermacol. 19, 117e125.
Ambient. 8, 15e27. Shishehbor, M.H., Brennan, M.L., Aviles, R.J., Fu, X., Penn, M.S., Sprecher, D.L.,
Graf, U., Van Schaik, N., 1992. Improved high bioactivation cross for the wing so- Hazen, S.L., 2003. Statins promote potent systemic antioxidant effects through
matic and recombination test in Drosophila melanogaster. Mutat. Res. 271, specific inflammatory pathways. Circulation 108, 426e431.
59e67. Sleijfer, S., Van Der Grasst, A., Planting, A.S., Stoter, G., Verweij, J., 2005. The potential
P.C. Orsolin et al. / Food and Chemical Toxicology 90 (2016) 10e17 17

of statins as part of anti-cancer treatment. Eur. J. Cancer 41, 516e522. vascular endothelial cells via the inhibition of the geranylgeranylation of RhoA.
Sparrow, C.P., Burton, C.A., Hernandez, M., Mundt, S., Hassing, H., Patel, S., Rosa, R., Life Sci. 79, 1484e1492.
Hermanowski-Vosatka, A., Wang, P.R., Zhang, D., Peterson, L., Detmers, P.A., Tijet, N., Helvig, C., Feyereisen, R., 2001. The cytochrome P450 gene superfamily in
Chao, Y.S., Wright, S.D., 2001. Simvastatin has anti-inflammatory and anti- Drosophila melanogaster: annotation, introneexon organization and phylogeny.
atherosclerotic activities independent of plasma cholesterol lowering. Arterio- Gene 262, 189e198.
sclerosis Thrombosis Vasc. Biol. 21, 115e121. Ungureanu, D., Filip, C., Artenie, A., Artenie, R., 2003. Evaluation of simvastatin
Spindler, R.S., Li, R., Dhahbi, J.M., Yamakawa, A., Mote, P., Bodmer, R., Ocorr, K., antioxidant effects. Rev. Med. Chir. Soc. Med. Nat. Iasi 107, 66e71.
Willians, R.T., Wang, Y., Ablao, K.P., 2012. Statin treatment increases lifespan and Yamashita, M., Otsuka, F., Mukai, T., Otani, H., Inagaki, K., Miyoshi, T., Goto, J.,
improves cardiac health in Drosophila by decreasing specific protein pre- Yamamura, M., Makino, H., 2008. Simvastatin antagonizes tumor necrosis
nylation. PLoS One 7 (6), e39581. factor-a inhibition of bone morphogenetic proteins-2-induced osteoblast dif-
Takeda, I., Maruya, C.I., Shirasaki, T., Misukami, H., Takahata, T., Myers, J.N., ferentiation by regulating Smad signaling and Ras/Rho-mitogen-activated
Kakehata, S., Yagihashi, S., Shimkawa, H., 2007. Simvastatin inactivates b1- protein kinase pathway. J. Endocrinol. 196, 601e613.
integrin and extracellular signal-related kinase signaling and inhibits cell pro- Yanae, M., Tsubaki, M., Satou, T., Itoh, T., Imano, M., Yamazoe, Y., Nishida, S., 2011.
liferation in head and neck squamous cell carcinoma cells. Cancer Sci. 98, Statin induced apoptosis via the suppression of ERK1/2 and Akt activation by
890e899. inhibition of the geranylgeranyl-pyrophosphate biosynthesis in glioblastoma.
Tang, D., Park, H., Georgescu, S.P., Sebti, S.M., Hamilton, A.D., Galper, J.B., 2006. J. Exp. Clin. Cancer Res. 30, 74e82.
Simvastatin potentiates tumor necrosis factor a-mediated apoptosis of human

Você também pode gostar