You are on page 1of 10

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/51552309

Molecular cytogenetic evaluation of the aneugenic effects of teniposide in


somatic and germinal cells of male mice

Article  in  Mutagenesis · August 2011


DOI: 10.1093/mutage/ger051 · Source: PubMed

CITATIONS READS

10 73

1 author:

Sabry Attia
King Saud University
179 PUBLICATIONS   2,966 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

IQGAP1 RNAi View project

All content following this page was uploaded by Sabry Attia on 10 September 2015.

The user has requested enhancement of the downloaded file.


Mutagenesis Advance Access published August 6, 2011
Mutagenesis pp. 1–9, 2011 doi:10.1093/mutage/ger051

Molecular cytogenetic evaluation of the aneugenic effects of teniposide in somatic and


germinal cells of male mice

Sabry M. Attia* this enzyme is an ideal target for cancer chemotherapeutic


Department of Pharmacology, College of Pharmacy, King Saud University, PO
drug (3). Virtually, every form of cancers that can be cured by
Box 11451 Riyadh, Saudi Arabia. systemic chemotherapy were always treated with regimens that
focus on (or at least contain) drugs targeted to topoisomerase
*
II (4). Indeed, drugs targeted to topoisomerase II are now
To whom correspondence should be addressed. Tel: þ966 542927708; Fax:
þ966 14677200; Email: attiasm@yahoo.com or attiasm@ksu.edu.sa
indispensable components of systemic chemotherapy schedule,
which could effectively cure several known types of human
Received on April 25, 2011; revised on June 18, 2011; cancers.
accepted on July 2, 2011 Teniposide is a podophyllotoxin derivative and was found to
The ability of topoisomerase II inhibitor, teniposide, to be able to increase the concentration of topoisomerase II–DNA
induce aneuploidy and meiotic delay in somatic and cleavage complexes (3). This action converts topoisomerase
germinal cells of male mice was investigated by fluores- II to a potent cellular toxin that fragments the genome.
cence in situ hybridisation (FISH) assay using labelled Consequently, teniposide is referred to as a topoisomerase II
DNA probes and 5-bromo-2#-deoxyuridine (BrdU) in- poison. It has been known for more than a decade that

Downloaded from mutage.oxfordjournals.org at King Saud University on September 3, 2011


corporation assay, respectively. Colchicine and mitomycin teniposide stabilises topoisomerase II-associated double-
C were used as a positive control aneugen and clastogen, stranded DNA breaks by inhibiting the ability of the enzyme
respectively, and these compounds produced the expected to ligate cleaved nucleic acid molecules (4). A pervious study
responses. Using FISH assay with centromeric and that examined drug effects on human topoisomerase IIa
telomeric DNA probes for erythrocyte, micronuclei (MN) indicated that podophyllotoxins action at both scissile bonds
showed that teniposide is not only clastogenic but also are required to stabilise double-stranded breaks in the genetic
aneugenic in somatic cells in vivo. The assay also showed material (5). The presence of podophyllotoxins at either scissile
that chromosomes can be enclosed in the MN before and bond inhibits DNA ligation only on that strand and hence
after centromere separation. By using the BrdU incorpo- stabilises a single-stranded break in the double helix (5). It has
ration assay, it could be shown that the meiotic delay been shown that in the treatment against small cell lung cancer,
caused by teniposide in germ cells was 48 h. Disomic and malignant lymphoma, breast cancer and ovarian cancer etc.
diploid sperms were shown in epididymal sperm hybri- through both clinical findings from chemotherapy results and in
dised with DNA probes specific for chromosomes 8, X and vitro chemosensitivity test of tumour specimen, anti-tumour
Y after teniposide treatment. The prevalence of autodiploid effect on teniposide was stronger than other chemotherapeutic
(XX88 and YY88) sperm and disomic XX8 or YY8 sperm drugs (6,7). However, podophyllotoxins utilisation is associ-
indicates that the second meiotic division was more ated with an increased risk of secondary acute myeloid
sensitive to teniposide than the first meiotic division. The leukaemia and increased sperm frequencies with chromosomal
results also suggest that earlier prophase stages contribute abnormalities (8,9).
relatively less to teniposide-induced aneuploidy. Both the As with many other anticancer drugs, high doses of
clastogenic and the aneugenic potential of teniposide can podophyllotoxins as etoposide and teniposide induce apoptosis
give rise to the development of secondary tumours and and disruption of inner mitochondrial membrane potential
abnormal reproductive outcomes in cured cancer patients (10,11). However, several lines of evidence indicate that low
and medical personnel exposing to drug regimens that doses of various chemotherapeutic drugs are capable of inducing
include teniposide. Thus, genetic counselling of these mitotic catastrophe, resulting from abnormal mitotic events that
patients should take place before the start of chemotherapy
produce improper chromosomal segregation and cell division,
and should take the present results into consideration.
leading to the formation of cells bearing mutations (12). These
cells can develop resistance to the therapeutic agents or may lead
to the development of secondary tumours and abnormal
reproductive outcomes. Chromosomal imbalance (aneuploidy)
Introduction induced by aneugens is a phenomenon observed in .90% of all
Type II topoisomerases are essential nuclear enzymes found in solid tumours. Even haematological cancers, known to have
all living organisms (1). Their basic role in cells is to catalyse a rather stable chromosome number, frequently display loss or
the transport of one DNA double helix through a transient gain of few chromosomes (13). Diploidy and aneuploidy in
double-strand break in another DNA molecule. This activity germ cells may cause abortions or children with congenital
helps relieve tensions built up in DNA during various DNA abnormalities such as trisomy 21, Klinefelter’s or Turner
metabolic processes, such as DNA replication, chromosome syndromes, respectively (14).
condensation and decondensation, chromosome segregation Teniposide has been reported to exert strong mutagenic
and transcription (2). Because of its essential role in cell growth, effects in mammalian cells in vitro but not in bacteria (15–17).
cell cycle, as well as the high expression in proliferating cells, It is a potent clastogenic agent in mouse lymphoma cell lines
Ó The Author 2011. Published by Oxford University Press on behalf of the UK Environmental Mutagen Society.
All rights reserved. For permissions, please e-mail: journals.permissions@oup.com. 1
S. M. Attia

(17) and it has been reported to induce sister chromatid Bone marrow FISH analysis of MN using centromeric and telomeric DNA probes
exchanges in CHO cells (18). Exposure of V79 cells to dif- Two DNA probes were used for tandem labelling: a murine minor satellite
ferent concentrations of teniposide resulted in a concentration- DNA probe specific for the centromeric region (27) and a murine hexamers
DNA probe specific for the telomeric region (28). The centromeric and
related elevation in the frequency of micronucleated binucleate telomeric probes were labelled with the fluorescent dyes Cy3 and fluorescein-
cells (19). Teniposide induced chromosomal aberrations, isothiocyanate (FITC) using the biotin or digoxygenin systems according to the
aneuploidy and micronucleus formation in CHO cells (20,21). manufacturer’s instructions (GIBCO Invitrogen, Carlsbad, CA, USA). In situ
The drug also induced sister chromatid exchange in V79 cells hybridisation assay was performed as described elsewhere (29–32) with slight
modifications. Briefly, frozen slides were brought to room temperature and
and mutation and somatic recombination in Drosophila rehydrated in 2 saline sodium citrate buffer (SSC) buffer for 5 min prior to
melanogaster in the wing spot test (22,23). Despite podo- treatment with 0.1% Triton X-100 in 2 SSC for 3 min. After washing the
phyllotoxin’s increasing use in malignancies, scarce data are slides in 2 SSC for 2 min, they were fixed in 4% paraformaldehyde for 10
available in literature on its potential aneugenicity in vivo. In min at room temperature and washed again in 2 SSC for 3 min. Then, the
an effort to predict patients’ reaction to tumour therapy, the slides were denaturated for 10 min in 70% formamide at 78°C and dehydrated
in an ice-cold ethanol series of 70, 90 and 100% for 2 min each. After air-
present study was designed to evaluate the aneugenicity of drying, the slides were brought to 37°C for ,3 min on a slide warming plate
teniposide in somatic and germinal cells of male mice in vivo. prior to mixing with the denaturated hybridisation mix. The hybridisation mix
The bone marrow micronucleus test complemented by contained 20 parts of the master mix 1.0 (50% formamide and 10% dextran
fluorescence in situ hybridisation (FISH) assay was performed sulphate), 2 parts of centromeric probe (20 ng per slide), 2 parts of telomeric
probe (12 ng per slide), 3 parts of herring sperm DNA (500 lg/ml) and 3
in mouse bone marrow cells to determine the aneugenic and parts of distilled water. The hybridisation mix was denatured for 5 min at 78°C
clastogenic origin of induced micronuclei (MN). The 5-bromo- and chilled on ice. Aliquots of 30 ll of the hybridisation mix were added to
2#-deoxyuridine (BrdU) incorporation assay was used to test if each slide, covered with coverslip and sealed with rubber cement. Hybridisation
teniposide treatment altered the duration of the meiotic was carried out overnight in a moist chamber at 37°C.
divisions and the sperm-FISH assay was performed for Post-hybridisation washing consisted of two steps: 40 min in 50%
formamide at 45°C and two washes for 15 min at 37°C in phosphate-nonidet
aneugenicity induction during male meiosis. In order to P-40 (PN) buffer (0.1 M sodium phosphate, 0.1% Nonidet P40, pH 8.0). After
determine the reliability of the methods, two model mutagens, incubation with 40 ll PNBR (PN buffer plus 5% blocking reagent and 0.02%

Downloaded from mutage.oxfordjournals.org at King Saud University on September 3, 2011


colchicine and mitomycin C, known to be predominantly sodium azide) for 10 min, the hybridised probes were detected by streptavidin-
aneugenic and clastogenic, respectively, were used as positive Cy3 for the biotin-labelled centromeric DNA probe and anti-digoxigenin–FITC
for the digoxigenin-labelled telomeric DNA probe. The hybridisation signals
control substances. were amplified by two rounds of incubation with biotinylated anti-streptavidin
followed by streptavidin-Cy3 and FITC-labelled anti-sheep IgG antibody made
in rabbit. The cells were counterstained with 4,6-diamidino-2-phenylindole
Materials and methods [DAPI; 0.1–0.5 lg/ml in phosphate-buffered saline (PBS)] for 10 min at room
temperature and coverslipped in Vectashield mounting medium. The slides
Animals
were scored immediately or following storage for several days at 4°C in the
Adult male white Swiss albino mice weighing 23–27 g (10–12 weeks old) were dark. The two applied DNA probes are identified by the regular appearance of
obtained from Experimental Animal Care Center at our university. The animals a red domain for centromeric DNA probe (labelled with Cy3) and a green
were maintained in an air-conditioned animal house at a temperature of 25– domain for the telomeric DNA probe (labelled with FITC). Strict scoring
28°C, relative humidity at 50% and photocycle of 12:12 h light and dark criteria were applied to avoid misinterpretation of signal numbers. MN were
periods. The animals were provided with standard diet pellets and water ad scored as having two or more domains of the same colour if the signals were
libitum. All experiments were carried out according to the Guidelines of the (i) nearly similar in size and intensity, (ii) separated by at least the distance of
Animal Care and Use Committee at our university. Each treatment group and half the diameter of the domain and (iii) regular in appearance, not diffuse and
vehicle control group consisted of five randomly assigned animals. The total clearly positioned within the MN. For signal detection, 50–150 MN per group
number of animals in the somatic cell study was 15 treated mice and 5 vehicle were examined for the presence or absence of DNA probes and the numbers of
control whereas the total number of animals in the germ cell studies was 60 signals per MN were counted under an Axioplan Fluorescence Microscope.
treated and 35 vehicle control mice.
Sperm BrdU incorporation assay
Drugs and treatment The time of development from meiotic divisions in spermatocytes to epididymal
Teniposide (Developmental Therapeutics Program, National Cancer Institute, sperm was assessed by labelling cells with BrdU. Mice were injected with 100 mg/
Bethesda, MD, USA) was dissolved in dimethyl sulfoxide (DMSO) in sterile kg BrdU in order to label spermatocytes at S-phase during preleptotene of meiosis.
dH2O, mixed on a magnetic stirrer for at least 30 min prior to administration During meiosis I and II, 13 days later, the mice were treated with 24 mg/kg
and administered by intraperitonial injection within 1 h following preparation. teniposide. For human chemotherapy, teniposide is administered typically at doses
Colchicine and mitomycin C (Sigma Chemical St Louis, MO, USA) were of 30–50 mg/m2/day for 5 days or three doses of 200 mg/m2 over 7 days. The
dissolved in sterile water and were used as a positive control aneugen and mouse has a body weight/surface area ratio of 3 kg/m2. Thus, the dose of 24 mg/
clastogen, respectively. Control mice receiving equal amount of DMSO (15% kg used in the current study corresponds to 72 mg/m2 and is within the dose range
DMSO in single treatment or 2% in multiple treatment) was included in order used for human chemotherapy. Five treated and five solvent control mice were
to code the slides and avoid scoring biases. The injected volume was 0.1 ml/10 g sacrificed per day 33–37 after BrdU injection (20–24 days after drug treatment).
body weight. All other chemicals were of the finest analytical grade. Both Caudae epididymes from each male were isolated and several incisions
were made. The epididymes were then placed in eppendorf cups with 300 ll of
Bone marrow conventional micronucleus test foetal calf serum and the sperms were allowed to actively leave the epididymes for
Animals were treated with 0.5 mg/kg teniposide and bone marrow was sampled 60 min at 37°C. Sperm suspensions (7 ll) were pipetted onto one end of a clean
24 h after treatment. The dose of teniposide was selected based on the data of dry glass slide and spread across the slide (33). Decondensation of the sperm
Bakheet et al. (24). Colchicine and mitomycin C were used as a positive control heads was performed by incubation of the coded slides in 10 mM dithiothreitol
aneugen and clastogen, respectively, at the dose of 2 mg/kg each (25). Bone for 30 min on ice followed by incubation in 4 mM lithium 3,5-diiodosalicylic acid
marrow smears were prepared and stained with May-Gruenwald/Giemsa for 60 min at room temperature. The slides were denatured in 2N HCl at room
solutions (26). At least four slides were made for each animal and allowed to temperature for 60 min and washed two times for 10 min in a Coplin jar with PBS
dry overnight. One slide per animal was stained with May-Gruenwald/Giemsa at room temperature to neutralise the acid. Immunodetection was carried out in
solutions for conventional assessment of the MN frequencies in polychromatic two steps in a humidified chamber: slides were incubated with 40 ll per slide of
erythrocytes (PCEs) and normochromatic erythrocytes (NCEs). The remaining a rat anti-BrdU (Seralab, 1/100) antibody for 30 min at room temperature. After
unstained slides were stored at 20°C for the distinction between the washing for 10 min with PN buffer at room temperature, the slides were
clastogenic and aneugenic effects by identifying the origin of MN with the incubated with 40 ll per slide of a FITC-conjugated anti-rat IgG antibody
mouse DNA probes. Per animal, 1000 PCE of coded slides were scored for the (Dianova, 1/100) for 30 min at room temperature and washed again for 10 min in
presence of MN. In addition, the number of PCEs among 1000 NCE per animal PN buffer at room temperature. Propidium iodide (2 lg/ml) was used as
was recorded to evaluate bone marrow suppression and mitotic activity was counterstaining. Averages of 10 000 BrdU-positive and BrdU-negative cells per
calculated as %PCE 5 [PCE/(PCE þ NCE)]  100. animal were blind scored under a Fluorescence Microscope.

2
Aneugenic effects of teniposide

Sperm multicolour FISH assay of bone marrow was noted (P , 0.05). In NCEs, the MN
Random groups of five mice each were treated with single doses of 6, 12 and 24 frequencies were between 0.02 and 0.06/100 NCE in all
mg/kg teniposide. Colchicine was used as a positive control aneugen at the dose groups. Thus, no discrimination between MN induced in PCE
of 3 mg/kg (34). After drug administration, the animals were maintained with
food and water ad libitum until being sacrificed. Mice were sacrificed by and NCE was required for the fluorescent analysis of MN with
cervical dislocation 23 days after drug treatment. In order to determine if in situ hybridisation since the NCE only contributed minimally
subacute treatment would have an effect because prophase stages would be to the total number of MN.
included in teniposide treatment, doses of 0.5, 1 and 2 mg/kg of teniposide in
2% DMSO were injected on 12 consecutive days and sperms were sampled 23 Bone marrow FISH analysis of MN using centromeric and
days after the last treatment. Immediately after animal sacrificing, the sperm
were collected from the C. epididymis. The time of sampling was chosen on the
telomeric DNA probes
basis of the BrdU incorporation study. The results for double labelling with centromeric and telomeric
The frequencies of disomic and diploid sperm were determined by FISH probes in both PCE and NCE are shown in Table II. The
with DNA probes specific for mouse chromosomes 8 (35), X (36) and Y (37). telomere-positive MN without a centromere signal represents
The probes for chromosomes 8 and Y were labelled with the fluorescent dyes
Cy3 and FITC using the biotin or digoxygenin systems, respectively, while the MN containing an acentric fragment with pericentric hetero-
chromosome X probe was labelled with a combination of Cy3 and FITC using chromatin. All MN with centromeric signals also had telomeric
the biotin and digoxygenin systems according to the manufacturer’s signals and these MN contained entire chromatids or entire
instructions (GIBCO Invitrogen, Carlsbad, CA, USA). The probe mixture chromosomes. After treatment of mice with the positive control
[4.5 lg probes in 2.1. master mix (55% formamide, 10% dextran sulfate) 2
SSC] was denatured at 78°C for 10 min and chilled on ice. Hybridisations with
aneugen colchicine, the majority of induced MN were
fluorochrome-labelled DNA probes were performed as previously described by telomere- as well as centromere positive, reflecting the
Attia et al. (34) with some modifications. Briefly, the sperm heads were expected aneugenic effects of colchicine. After treatment of
decondensed as described above in BrdU incorporation assay and then the mice with the positive control clastogen mitomycin C, only
slides were denaturated for 10 min in 70% formamide at 78°C and dehydrated 19.27% of induced MN were double-labelled with the telomere
in ethanol series. Following the application of 30 ll of probe mixture,
hybridisation was performed at 37°C for 24–48 h in a moist chamber. Post- and the centromere probes, confirming the predominantly
hybridisation washing and detection of the hybridised probes were performed clastogenic effects of mitomycin C. As shown in Figure 1 and

Downloaded from mutage.oxfordjournals.org at King Saud University on September 3, 2011


as described above in bone marrow FISH analysis. Sperm nuclei were Table II, the ratios of MN arising from clastogenic (centromere
counterstained with 0.05–0.1 lg/ml DAPI. Slides were examined for disomic negative) and aneugenic (centromere positive) effects of the
and diploid sperm under an Axioplan Fluorescence Microscope. Per animal,
fluorescent signals from coded slides were counted in 10 000 sperms and
vehicle and positive controls were found to be in the same
sperms were designated as normal (X8 and Y8), disomic (X88, Y88 and XY8) range as reported earlier (29–32). With teniposide, 18.24% of
or diploid (XY88, XX88 and YY88). the analysed MN had no signal, 35.14% showed telomere
signals and 46.62% were double labelled with the telomere and
Statistical analysis
the centromere probes. The distribution of the signal
Data were expressed as the mean  standard deviation (SD) of the means. The
analysis parameters were tested for homogeneity of variance and normality and frequencies in MN that were positive only with the telomere
were found normally distributed. The data were, therefore, analysed by probe is shown in Table III. The majority of analysed MN in
employing non-parametric tests, the Mann–Whitney U-test. Results were
considered significantly different if the P value was 0.05.

Table II. Results of double FISH labelling of colchicine (2 mg/kg),


Results mitomycin C (2 mg/kg) and teniposide (0.5 mg/kg)-induced MN with the
telomeric and the centromeric DNA probes
Bone marrow conventional MN test
The incidences of micronucleated polychromatic erythrocyte Chemicals In situ hybridisation analysis of MN
(MNPCE) in each treatment group as well as the PCE/NCEs Number MN MN with MN with
ratio are shown in Table I. After treatment with the positive of MN without any telomere telomere and
controls colchicine and mitomycin C, a statistically significant scored signal (%) signals centromere
increase in the incidence of MNPCE over the control value was only (%) signals (%)
observed (P , 0.01). Moreover, mitomycin C significantly Control 53 16 (30.19) 14 (26.42) 23 (43.39)
decreased the percent PCE (P , 0.05), indicating a reduction Colchicine 80 9 (11.25) 13 (16.25) 58 (72.5)
in erythroblast proliferation. Teniposide significantly increased Mitomycin C 83 14 (16.87) 53 (63.86) 16 (19.27)
MNPCE frequencies (P , 0.01) and the response was directly Teniposide 148 27 (18.24) 52 (35.14) 69 (46.62)
correlated with bone marrow toxicity as significant suppression

Table I. Frequencies of MNPCE, MNNCE and %PCE in bone marrow of


mice after treatment with colchicine (2 mg/kg), mitomycin C (2 mg/kg) and
teniposide (0.5 mg/kg)

Chemicals % MNPCE % MNNCE % PCE


(mean  (mean  (mean 
SD) SD) SD)

Control 0.30  0.07 0.02  0.04 48.6  2.60


Colchicine 1.06  0.16** 0.02  0.04 45.4  3.78
Mitomycin C 1.33  0.34** 0.04  0.05 41.6  3.43*
Teniposide 3.88  0.57** 0.06  0.05 42.8  4.32*
Fig. 1. Illustration of the contribution of clastogenicity and aneugenicity to the
*P , 0.05, **P , 0.01 compared with the solvent corresponding control induced MN frequencies in animals treated with colchicine (2 mg/kg),
(Mann–Whitney U-test). mitomycin C (2 mg/kg) and teniposide (0.5 mg/kg).

3
S. M. Attia

colchicine group contained one telomere signal, whereas the groups were significantly below the control values (P , 0.05
majority of analysed MN in mitomycin C contained four and P , 0.01, respectively), while on days 23 and 24, there were
telomere signals. Of the 52 MN analysed following treatment no significant differences between the teniposide-treated and
of mice with the teniposide, one telomere signal per MN was control animals. According to Oakberg (38), the development
observed in 38.46% MN, two signals were seen in 32.69% from meiotic spermatocytes of mice to epididymal sperm takes
MN, three signals were seen in 17.3% MN and four signals 22 days. Thus, the meiotic delay caused by teniposide was 48
were seen in 11.53% MN. h. Therefore, the optimum day for sperm sampling in the sperm-
The distribution of the signal frequencies in the MN that were FISH assays was concluded to be 23 or 24 with teniposide.
double labelled with both the centromere and the telomere
probes is shown in Table IV. The majority of analysed MN in Multicolour sperm-FISH assay
colchicine group contained one centromere and two telomere The results of the analysis of aneugenic effects in germ cells of
signals or two centromere and four telomere signals. After male mice after single exposure to teniposide are presented in
treatment with mitomycin C, only 25.0% double-labelled MN Table V, together with the negative and positive control data.
had one centromere and one or two telomere signals, 31.25% Sex ratios were found to be in the same range as the theoretical
showed one centromere and three or four telomere signals, ratio of 1:1 for X- versus Y-bearing sperm in all groups. A
18.75% showed two centromere and one or two telomere signals significant increase in the frequency of diploid sperm was
and 25.0% showed two centromere and three or four telomere caused by treatment with all doses of teniposide compared with
signals. With teniposide, 40.5% of induced MN had one the control value. Treatment of mice with 6 mg/kg of
centromere and one or two telomere signals, 23.09% showed teniposide did not induce significant increases in disomic
one centromere and three or four telomere signals, 23.09% sperm; however, treatment with 12 and 24 mg/kg of teniposide
showed two centromere and one or two telomere signals and significantly induced disomic sperm compared with the control
17.38% showed two centromere and three or four telomere value. The dose–response curves for teniposide-induced
signals. To correlate the FISH data with the conventional MN disomic and diploid sperm can be described by the linear

Downloaded from mutage.oxfordjournals.org at King Saud University on September 3, 2011


data, the expected percent of PCE with signals-positive and equations y 5 0.004x þ 0.041 (r2 5 0.990) and y 5 0.002x þ
-negative MN were calculated (Figure 1). For example, after 0.003 (r2 50.998), respectively (Figure 3). The frequency of
treatment with colchicine, 1.06% MNPCE were found in the disomic sperm induced by colchicine was significantly in-
conventional MN test and 88.75% MN were signals positive in creased by a factor of 1.64 compared with the control value.
the FISH analysis; thus, 0.94075% of the 1.06% MNPCE were However, in contrast to teniposide, colchicine did not
calculated to be signals positive and, correspondingly, 0.11925% significantly increase the frequency of diploid sperm, in-
MN were calculated to be signals negative. dicating that no complete meiotic arrest occurred.
The results of the multiple exposures to teniposide are shown
Sperm BrdU incorporation assay in Table VI. There were no mortality or symptoms of toxicity
The results of the BrdU incorporation assay are presented in observed after repeated treatment with teniposide or the vehicle.
Figure 2. With 24 mg/kg of teniposide, prolongations of the These results indicate that repeated treatment with teniposide and
duration of the meiotic divisions were observed. On days 21 and the vehicle are well tolerated by experimental animals. Treatment
22, the frequencies of BrdU-labelled sperm in the teniposide of mice with 0.5 and 1 mg/kg of teniposide on 12 consecutive
days did not induce any significant increases of disomic or
diploid sperm. However, the highest dose group (2 mg/kg/day),
which received a total of 24 mg/kg, induced significant increases
Table III. Distribution of the telomere frequencies among MN after treatment
with colchicine (2 mg/kg), mitomycin C (2 mg/kg) and teniposide (0.5 mg/
in disomic and diploid sperm compared with the corresponding
kg) control values. The dose–response curves for teniposide-induced
disomic and diploid sperm can be described by the linear
Chemicals MN with One Two Three Four equations y 5 0.017x þ 0.048 (r2 5 0.838) and y 5 0.011x þ
telomere telomere telomere telomere telomere
signals signal (%) signals (%) signals (%) signals (%)
0.001 (r2 50.936), respectively (Figure 4).
only

Control 14 8 (57.14) 3 (21.42) 2 (14.28) 1 (7.14) Discussion


Colchicine 13 7 (53.84) 3 (23.07) 1 (7.692) 2 (15.38)
Mitomycin C 53 10 (18.86) 12 (22.64) 14 (26.41) 17 (32.07) The genotoxic activity of teniposide during mitosis in vivo was
Teniposide 52 20 (38.46) 17 (32.69) 9 (17.30) 6 (11.53) recently confirmed in our laboratory with the bone marrow MN

Table IV. Distribution of the signal frequencies in the double FISH labelled MN with the centromere and the telomere DNA probes after induction with colchicine
(2 mg/kg), mitomycin C (2 mg/kg) and teniposide (0.5 mg/kg)

Chemicals Number One One One One Two Two Two Two
of double- centromere centromere centromere centromere centromere centromere centromere centromere
labelled and one and two and three and four and one and two and three and four
MN telomere telomere telomere telomere telomere telomere telomere telomere
signals (%) signals (%) signals (%) signals (%) signals (%) signals (%) signals (%) signals (%)

Control 23 6 (26.08) 3 (13.04) 2 (8.69) 3 (13.04) 3 (13.04) 2 (8.69) 3 (13.04) 1 (4.34)


Colchicine 58 3 (5.17) 10 (17.2) 2 (3.44) 8 (13.79) 7 (12.06) 8 (13.79) 8 (13.79) 12 (20.6)
Mitomycin C 16 3 (18.75) 1 (6.25) 3 (18.75) 2 (12.5) 2 (12.5) 1 (6.25) 2 (12.5) 2 (12.5)
Teniposide 69 12 (17.4) 16 (23.1) 6 (8.69) 10 (14.4) 8 (11.59) 5 (7.24) 4 (5.79) 8 (11.59)

4
Aneugenic effects of teniposide

test (24). So far the origin of the teniposide-induced MN is not arrest. A total of 148 MN from teniposide-treatment group
determined yet and there are no in vivo published aneuploidy were analysed by FISH and 69 (46.62%) MN of these were
studies for teniposide in germ cells. Therefore, the current double labelled with centromere and telomere probes, in-
study was designed to investigate the ability of teniposide to dicating that they were formed by lagging chromosomes and
induce aneuploidy in somatic and germinal cells of male mice. reflecting an aneugenic activity of teniposide. Similarly, 77
The conventional bone marrow MN test complemented by (53.38%) of the induced MN were centromere negative,
FISH with the mouse centromeric and telomeric DNA probes indicating that they were formed by acentric fragments and
was used to investigate the mechanisms of induction of MN in reflecting the clastogenic activity of teniposide. Since the
mice treated with teniposide. This assessment is particularly centromere probe labelled mitotic chromosomes at both
important for chemical clastogens, which may also have chromatids, it can be concluded that equal proportions of
aneugenic potential. Such chemicals may interact, on the MN contained single chromatids (chromosome loss) and
chromosomal level, with the respective targets for chromo- biarmed chromosomes (non-disjunction).
somal missegregation. To determine the efficiency of the These results indicate that teniposide is clastogenic and
method, two model mutagens, colchicine and mitomycin C, aneugenic during mitotic phases. Both the clastogenic and
known to have aneugenic and clastogenic actions, respectively, the aneugenic potential of teniposide in somatic cells can give
were used. The observed results of the MNPCE and the rise to the development of secondary tumours. Therefore, the
distribution of signals per MN in the solvent control and the clinical use of this genotoxic drug must be weighed against
two positive mutagens in the current study correspond well the risks of secondary malignancies in cured patients.
with the published data reported earlier (29,32). These data Importantly, experimental observations have shown that the
confirmed the sensitivity of the experimental protocol followed catalytic topoisomerase II inhibitor, dexrazoxane, is able to
in the detection of genotoxic effects. enhance the cytotoxic action of teniposide (39) and modulates
Teniposide at a dose of 0.5 mg/kg significantly increased the teniposide-induced DNA damage and apoptosis in the bone
frequency of MNPCE, as expected (24). Additionally, tenipo- marrow cells in vivo (24). Thus, based on the results of these

Downloaded from mutage.oxfordjournals.org at King Saud University on September 3, 2011


side significantly decreased the percent PCE, indicating previous in vivo studies, strategies can be developed to
a reduction in erythroblast proliferation most likely by mitotic decrease the teniposide-induced genotoxicity in non-tumour
cells using dexrazoxane. A total of 35.14% of the analysed
MN was telomere positive and must be interpreted as MN
harbouring terminal acentric fragments (Table III). Addition-
ally, 18.24% of the analysed MN had no FISH signals and
must be interpreted as MN harbouring interstitial acentric
fragments (Table II). Centromere plus telomere double-
labelled MN that gave one centromeric signal and two
telomere signals (Table IV) probably contained a chromosome
lagging after centromere separation. MN with two centromere
signals and four telomere signals most likely contained
a chromosome lagging before centromere separation or two
chromatids after centromere separation.
Studies in humans have shown that certain chemotherapy
regimens increase the frequencies of aneuploidy in germinal
cells (40–43), suggesting that such patients may be at higher
Fig. 2. Time course of appearance of BrdU-labelled sperm in the epididymis risk for the development of secondary tumours and abnormal
after treatment with 24 mg/kg teniposide. *P , 0.05, **P , 0.01 compared reproductive outcomes. Therefore, it is of general concern to
with the concurrent control (Mann–Whitney U-test).

Table V. Results of the multi-colour FISH with epididymal sperm of mice treated with colchicine and teniposide

Control Colchicine (3 mg/kg) Teniposide (6 mg/kg) Teniposide (12 mg/kg) Teniposide (24 mg/kg)

Number of animals 5 5 5 5 5
Sperm scored 50011 50029 50081 50012 50041
X/Y-bearing sperm 0.9984 1.0011 1.0515 1.0311 1.0081
Disomic sperm
X-X-8 6 14 9 11 18
Y-Y-8 7 12 7 11 17
X-Y-8 2 2 3 4 7
X-8-8 4 6 7 13 13
Y-8-8 6 6 6 9 13
Total 25 40 32 48 68
% Disomies  SD 0.05  0.010 0.08** 0.012 0.064  0.015 0.096**  0.015 0.136**  0.019
Diploid sperm
X-Y-8-8 0 0 2 3 5
X-X-8-8 1 1 5 9 16
Y-Y-8-8 0 1 3 5 11
Total 1 2 10 17 32
% Diploidies  SD 0.002  0.004 0.004  0.005 0.02  0.007* 0.034**  0.015 0.064**  0.005

*P, 0.05, **P, 0.01 compared with the concurrent control (Mann–Whitney U-test).

5
S. M. Attia

decrease the risk of aneuploidy production, detection of germ In the present experiments, teniposide caused dose-dependent
cell aneugens and understandings of the causal mechanisms are significant increases in the frequencies of disomic and diploid
essential. In the current study, aneuploidy was determined in sperm. The induction of aneuploidy showed linear dose–
germinal cell by the sperm-FISH assay. In order to determine responses between 0 and 24 mg/kg of teniposide. This in vivo
the reliability of the methods, colchicine, known to be observation is in line with an earlier in vitro report on the CHO
predominantly aneugenic, was used as positive control sub- cells that teniposide induces the formation of quadriradial
stance and the results of the positive and negative control were chromosomes (21). Polyploidy induced by teniposide was also
in the same range as those of the earlier studies (32,34,44,45). demonstrated by flow cytometry techniques in murine eryth-
These data confirmed the sensitivity of the experimental roleukemic cells (48). Furthermore, Morse-Gaudio and Risley
protocol followed in the detection of aneuploidogenic effects. (51) reported that incubation of isolated spermatogenic cells
It has been often discussed that chemicals with aneugenic Xenopus laevis with teniposide led to dose-dependent induction
properties can alter the progression of cell division in both of DNA breaks in all spermatocytes and spermatid stages to
meiotic and mitotic cells (46,47). In the present study, the time nuclear elongation stages, as analysed by alkaline single-cell gel
of development from meiotic divisions in spermatocytes to electrophoresis. To determine if subacute treatment with low
epididymal sperm was assessed by the BrdU incorporation doses of teniposide would have an effect because earlier prophase
assay. The results clearly indicate that teniposide prolonged the stages would be included in teniposide treatment. Individual
duration of the meiotic divisions in mouse spermatocytes for doses of 0.5, 1 and 2 mg/kg of each compound were injected on
48 h. These observations therefore confirm previous results 12 consecutive days and sperms were sampled 23 days after the
that inhibition of topoisomerase II function during different last treatment with teniposide. It was found that a total of 24
phases of the cell cycle by teniposide slow down cell cycle mg/kg teniposide applied to the entire prophase of meiosis
progression and causes cells to arrest at the G2/M phase significantly increased disomic and diploid sperm frequencies,
(11,48,49). Such G2/M arrest has been proposed to be due to while a total dose of 6 or 12 mg/kg was negative. In contrast,
induction of G2 checkpoint machinery that allows damaged a single dose of 6 mg/kg teniposide applied to spermatocytes

Downloaded from mutage.oxfordjournals.org at King Saud University on September 3, 2011


DNA to be repaired before cells move to the next cell cycle during the first meiotic division (meiosis I; MMI)/the second
stage (50). meiotic division (meiosis II; MMII) gave a positive result. These
data suggest that earlier prophase stages contribute relatively less
to teniposide-induced aneuploidy in male germ cells.
The sperm-FISH assay for disomy or diploidy is capable of
detecting effects induced during both meiotic divisions and to
compare the sensitivity of both meiotic divisions (52,53). In the
current study, it was found that teniposide caused noticeable
increases above the control of autodiploid sperm (XX88 and
YY88). After treatment with teniposide, autodiploid sperm
resulting from arrest of MMII were more frequent than diploid
sperm resulting from arrest during MMI (XY88). Thus, the
second meiotic division was more sensitive to teniposide
treatment than the first meiotic division. The conclusion that
second meiotic divisions were more sensitive to teniposide
treatment than the first meiotic divisions is also supported by
the observed frequencies of sex chromosome disomic. Sperm
Fig. 3. Dose–response curves of the frequencies of abnormal sperm from mice with signals of XX8 or YY8 were more frequent than sperm
after treatment with teniposide. *P , 0.05, **P , 0.01 compared with the with signals of XY8.
concurrent control (Mann–Whitney U-test).

Table VI. Results of the multicolour FISH with epididymal sperm of mice treated with teniposide (daily exposure for 12 consecutive days)

Control Teniposide (0.5 mg/kg) Teniposide (1 mg/kg) Teniposide (2 mg/kg)

Number of animals 5 5 5 5
Sperm scored 50078 50072 50171 50012
X/Y-bearing sperm 1.0044 0.9912 0.9952 1.0049
Disomic sperm
X-X-8 7 9 5 11
Y-Y-8 4 8 7 10
X-Y-8 2 3 3 3
X-8-8 8 5 6 12
Y-8-8 5 4 7 8
Total 26 29 28 44
% Disomies  SD 0.052  0.017 0.058  0.008 0.056  0.015 0.088**  0.01
Diploid sperm
X-Y-8-8 0 1 2 2
X-X-8-8 1 1 2 6
Y-Y-8-8 0 1 1 5
Total 1 3 5 13
% Diploidies  SD 0.004  0.005 0.006  0.005 0.01  0.007 0.026**  0.005

*P, 0.05, **P, 0.01 compared with the concurrent control (Mann–Whitney U-test).

6
Aneugenic effects of teniposide

aneugens and the possibility of unique germ cell aneugens


should not be neglected.
DNA topoisomerase inhibitors have a clear tendency to
cause stranded DNA breaks, which primarily result in the
formation of centromere-negative MN (32). Similarly, the
demonstration that teniposide is effective topoisomerase
inhibitor suggests that teniposide elicit its clastogenic effects
through this mechanism. Teniposide metabolites could also
activate cells to enhance their intracellular production of
reactive oxygen species, of which the stable and diffusible
forms could damage nuclear DNA (56). If the cellular repair
mechanisms are overloaded, primary DNA damage may lead to
structural or numerical chromosomal aberration and eventually
Fig. 4. Dose–response curves of the frequencies of abnormal sperm from mice cause tumours (57). However, the induction of centromere-
23 days after the last treatment with teniposide (daily exposure for 12 positive MN by teniposide indicates that there may be another
consecutive days). **P , 0.01 compared with the concurrent control (Mann–
Whitney U-test).
mechanism through which teniposide can induce its genotoxic
effect, an observation that also underscores the importance of
using the FISH modification of the MN assay to determine the
The data of sperm study indicate that male cancer patients origin of the induced MN. The possible mechanism by which
receiving chemotherapy with teniposide may stand a higher teniposide may exert its aneugenic effect is through inhibition
risk of siring chromosomally abnormal offspring, i.e. with of topoisomerase II which could result in a mal-segregation of
Down, Klinefelter or Turner syndromes. The chemotherapy chromosomes during cell divisions. As mentioned in In-
with teniposide may also reduce the fertility of the patients. The troduction, topoisomerase II has been demonstrated to be

Downloaded from mutage.oxfordjournals.org at King Saud University on September 3, 2011


most sensitive period of spermatogenesis to aneuploidy necessary for the proper separation of sister chromatids during
induction is meiosis. In humans, the time of development cell divisions (2) with both non-disjunction and breakage
from meiosis in spermatocytes to mature sperm takes 40–57 occurring in its absence (58). Thus, teniposide may be able to
days (38) with some uncertainty about the retention of sperm in inhibit two key roles of topoisomerase II, its ability to properly
the epididymis. Baumgartner et al. (54) could show that the segregate newly replicated chromosomes as well as its function
aneuploidy rates in sperm of Hungarian men were elevated to religate transient double-stranded DNA breaks.
significantly 40–50 days after attempted suicide with diazepam In summary, by using FISH assay with centromeric and
and returned to control levels when sperm were sampled 100 telomeric DNA probes for erythrocyte MN, it could be shown
days after the intoxication. Therefore, the hazard for patients to that teniposide is not only clastogenic but also aneugenic in
foster a chromosomally unbalanced child is only elevated somatic cells in vivo. The assay also showed that chromosomes
during 3–4 months after the end of chemotherapy with can be enclosed in an MN before and after centromere
aneugenic drugs. Often, cancer patients facing a limited life separation. By using the BrdU incorporation assay, it could be
expectancy express a great desire to have a child. Genetic shown that the meiotic delay caused by teniposide was 48 h.
counselling of these patients should take place before the start With the sperm-FISH analysis, it could be shown that teniposide
of chemotherapy and should take the present results into induce aneuploidies during meiosis that result in disomic sperm
consideration. as well as complete meiotic arrest that results in diploid sperm.
The present mouse bone marrow MN studies showed that The prevalence of autodiploid (XX88, YY88) sperm and
exposure to 0.5 mg/kg teniposide yielded a significant increase disomic XX8 or YY8 sperm indicate that the second meiotic
in MNPCE. However, in the sperm-FISH assay, it was found division was more sensitive to teniposide than the first meiotic
that the lowest positive dose, which caused disomic or diploid division. The results suggest also that earlier prophase stages
sperm, was 6 mg/kg of teniposide. This observation suggests contribute relatively less to teniposide-induced aneuploidy. Both
that MN in bone marrow are induced at lower doses than the clastogenic and the aneugenic potential of teniposide can
disomies or diploidies in sperm; hence, the bone marrow is the give rise to the development of secondary tumours and abnormal
more sensitive tissue. It must of course be noted that the assays reproductive outcomes in cancer patients and medical personnel
measure different end-points. Chromosome loss and breakage exposing to drug regimens that include teniposide. The results of
are measured in the MN test, and non-disjunction is detected in the current study and our previous data base show that the
the sperm-FISH assay. Therefore, the present data confirm the aneuploidy assays such as the sperm-FISH assay and MN test
general paradigm of hazard assessment that the positive complemented by FISH with centromeric DNA probes have
outcome of the bone marrow MN test is an indicator of the been sufficiently validated to be recommended for the
genotoxic potential of a compound in germ cells. However, to assessment of aneugenic effects of chemicals.
quantify aneuploidy induced in germ cells is important for risk
assessment purposes. Taking into account the fundamental Funding
differences between the meiotic process and the mitotic
process, e.g. requirement for chromosome pairing, formation Deanship of Scientific Research at King Saud University
of chiasmata to allow recombination, prolonged duration of through the research group project No. (RGP-VPP-120).
meiosis in oocytes (55), it will always be necessary to confirm
the aneugenic potential of a chemical detected in in vitro by Acknowledgements
studies in somatic and in germinal cells in vivo. Theoretically,
the differences in cell biology may give rise to qualitative The author is grateful to Mr Müller, M. for assistance with certain techniques.
differences in response of germ cells and somatic cells to Conflict of interest statement: None declared.

7
S. M. Attia

References 26. Attia, S. M. (2007) The genotoxic and cytotoxic effects of nicotine in the
mouse bone marrow. Mutat. Res., 632, 29–36.
1. Wang, J. C. (1996) DNA topoisomerases. Annu. Rev. Biochem., 65, 27. Wong, A. K. C. and Rattner, J. B. (1988) Sequence organization and
635–692. cytological localization of the minor satellite of mouse. Nuclei Acid Res.,
2. Wang, J. C. (2002) Cellular roles of DNA topoisomerases: a molecular 16, 11645–11661.
perspective. Nat. Rev. Mol. Cell Biol., 3, 430–440. 28. Ijdo, J. W., Wells, R. A., Baldini, A. and Reeders, S. T. (1991) Improved
3. Walker, J. V. and Nitiss, J. L. (2002) DNA topoisomerase II as a target for telomere detection using a telomere repeat probe (TTAGGG)n generated by
cancer chemotherapy. Cancer Invest., 20, 570–589. PCR. Nucleic Acids Res., 19, 4780.
4. Burden, D. A. and Osheroff, N. (1998) Mechanism of action of eukaryotic 29. Schriever-Schwemmer, G. and Adler, I.-D. (1994) Differentiation of
topoisomerase II and drugs targeted to the enzyme. Biochim. Biophys. Acta, micronuclei in mouse bone marrow cells: a comparison between CREST
1400, 139–154. staining and fluorescent in situ hybridization with centromeric and
5. Bromberg, K. D., Burgin, A. B. and Osheroff, N. (2003) A two-drug model telomeric DNA probes. Mutagenesis, 9, 333–340.
for etoposide action against human topoisomerase IIa. J. Biol. Chem., 278, 30. Attia, S. M., Kliesch, U., Schriever-Schwemmer, G., Badary, O. A.,
7406–7412. Hamada, F. M. and Adler, I. D. (2003) Etoposide and merbarone are
6. van der Gaast, A. and Splinter, T. A. (1992) Teniposide (VM-26) in ovarian clastogenic and aneugenic in the mouse bone marrow micronucleus test
cancer: a review. Semin. Oncol., 19 (2 Suppl. 6), 95–97. complemented by fluorescence in situ hybridization with the mouse minor
7. Bepler, G. and O’Briant, K. (1998) In vitro chemosensitivity testing of satellite DNA probe. Environ. Mol. Mutagen., 41, 99–103.
human non-small cell lung cancer cell lines. Anticancer Res., 18, 31. Attia, S. M. (2007) Chromosomal composition of micronuclei in mouse
3181–3185. bone marrow treated with rifampicin and nicotine, analyzed by multicolor
8. Smith, M. A., Rubinstein, L., Anderson, J. R. et al. (1999) Secondary fluorescence in situ hybridization with pancentromeric DNA probe.
leukemia or myelodysplastic syndrome after treatment with epipodophyl- Toxicology, 235, 112–118.
lotoxins. Clin. Oncol., 17, 569–577. 32. Attia, S. M. (2009) Use of centromeric and telomeric DNA probes in in situ
9. Martin, R. H., Ernst, S., Rademaker, A., Barclay, L., Ko, E. and hybridization for differentiation of micronuclei induced by lomefloxacin.
Summers, N. (1999) Analysis of sperm chromosome complements before, Environ. Mol. Mutagen., 50, 394–403.
during, and after chemotherapy. Cancer Genet. Cytogenet., 108, 133–136. 33. Schmid, T. E., Attia, S., Baumgartner, A., Nuesse, M. and Adler, I. D.
10. Miciæ, M., Bihari, N., Jaksiæ, Z., Müller, W. E. and Batel, R. (2002) DNA (2001) Effect of chemicals on the duration of male meiosis in mice detected
damage and apoptosis in the mussel Mytilus galloprovincialis. Mar. with laser scanning cytometry. Mutagenesis, 16, 339–343.
Environ. Res., 53, 243–262. 34. Attia, S. M., Schmid, T. E., Badary, O. A., Hamada, F. M. and Adler, I. D.

Downloaded from mutage.oxfordjournals.org at King Saud University on September 3, 2011


11. Li, J., Chen, W., Zhang, P. and Li, N. (2006) Topoisomerase II trapping (2002) Molecular cytogenetic analysis in mouse sperm of chemically
agent teniposide induces apoptosis and G2/M or S phase arrest of oral induced aneuploidy: studies with topoisomerase II inhibitors. Mutat. Res.,
squamous cell carcinoma. World J. Surg. Oncol., 4, 41.
520, 1–13.
12. Roninson, I. B., Broude, E. V. and Chang, B. D. (2001) If not apoptosis,
35. Boyle, A. L. and Ward, D. (1992) Isolation and initial characterization of
then what? Treatment-induced senescence and mitotic catastrophe in tumor
a large repeat sequence element specific to mouse chromosome 8.
cells. Drug Resist. Updat., 4, 303–313.
Genomics, 12, 517–525.
13. Mitelman, F., Johansson, B. and Mertens, F. (eds) (2011) Mitelman Database
36. Disteche, C. M., Tantrvahi, U., Gandy, S., Eisenhard, M., Adler, I. D. and
of Chromosome Aberrations and Gene Fusions in Cancer. http://cgap.nci.
Kunkel, L. M. (1985) Isolation and characterization of two repetitve DNA
nih.gov/Chromosomes/Mitelman. (accessed August 1, 2011).
fragments located near the centromere of the mouse X chromosome.
14. Fritz, B., Aslan, M., Kalscheuer, V., Ramsing, M., Saar, K., Fuchs, B. and
Cytogenet. Cell Genet., 39, 262–268.
Rehder, H. (2001) Low incidence of UPD in spontaneous abortions beyond
37. Bishop, C. E. and Hatat, D. (1987) Molecular cloning and sequence
the 5th gestational week. Eur. J. Hum. Genet., 9, 910–916.
15. Nakanomyo, H., Hiraoka, M. and Shiraya, M. (1986) Mutagenicity tests of analysis of a mouse Y chromosome RNA transcript expressed in the testis.
etoposide and teniposide. J. Toxicol. Sci., 11, 301–310. Nucleic Acids Res., 15, 2959–2969.
16. Gupta, R. S., Bromke, A., Bryant, D. W., Gupta, R., Singh, B. and 38. Oakberg, E. F. (1956) Duration of spermatogenesis in the mouse and timing
McCalla, D. R. (1987) Etoposide (VP16) and teniposide (VM26): novel of stages of the cycle of the seminiferous epithelium. Am. J. Anat., 99,
anticancer drugs, strongly mutagenic in mammalian but not prokaryotic test 507–516.
systems. Mutagenesis, 2, 179–186. 39. Holm, B., Sehested, M. and Jensen, P. B. (1998) Improved targeting of
17. DeMarini, D. M., Brock, K. H., Doerr, C. L. and Moore, M. M. (1987) brain tumors using dexrazoxane rescue of topoisomerase II combined with
Mutagenicity and clastogenicity of teniposide (VM-26) in L5178Y/ supralethal doses of etoposide and teniposide. Clin. Cancer Res., 4,
TKþ/-3.7.2C mouse lymphoma cells. Mutat. Res., 187, 141–149. 1367–1373.
18. Singh, B. and Gupta, R. S. (1983) Mutagenic responses of thirteen 40. Genescà, A., Caballı́n, M. R., Miró, R., Benet, J., Bonfill, X. and
anticancer drugs on mutation induction at multiple genetic loci and on sister Egozcue, J. (1990) Human sperm chromosomes. Long-term effect of cancer
chromatid exchanges in Chinese hamster ovary cells. Cancer Res., 43, treatment. Cancer Genet. Cytogenet., 46, 251–260.
577–584. 41. Robbins, W. A., Meistrich, M. L., Moor, D., Hagemeister, F. B., Weier, H.-U.,
19. Adiga, S. K. and Jagetia, G. C. (1999) Effect of teniposide (VM-26) on the Cassel, M. J., Wilson, G., Eskenazi, B. and Wyrobek, A. J. (1997)
cell survival, micronuclei-induction and lactate dehydrogenase activity on Chemotherapy induces transient sex chromosomal and autosomal aneuploidy
V79 cells. Toxicology, 138, 29–41. in human sperm. Nature Genet., 16, 74–78.
20. Albertini, S., Chételat, A. A., Miller, B., Muster, W., Pujadas, E., 42. Attia, S. M. (2008) Mutagenicity of some topoisomerase II-interactive
Strobel, R. and Gocke, E. (1995) Genotoxicity of 17 gyrase- and four agents. Saudi Pharm. J., 17, 1–24.
mammalian topoisomerase II-poisons in prokaryotic and eukaryotic test 43. Burrello, N., Vicari, E., La Vignera, S., Romeo, G., Campagna, C.,
systems. Mutagenesis, 10, 343–351. Magro, E., Giuffrida, D., D’Agata, R. and Calogero, A. E. (2011) Effects of
21. Charron, M. and Hancock, R. (1991) Chromosome recombination and anti-neoplastic treatment on sperm aneuploidy rate in patients with
defective genome segregation induced in Chinese hamster cells by the testicular tumor: a longitudinal study. J. Endocrinol. Invest., 29, e121–125.
topoisomerase II inhibitor VM-26. Chromosoma, 100, 97–102. 44. Attia, S. M., Badary, O. A., Hamada, F. M., de Angelis, M. H. and
22. Lim, M., Liu, L. F., Jacobson-Kram, D. and Williams, J. R. (1986) Adler, I. D. (2005) Orthovanadate increased the frequency of aneuploid
Induction of sister chromatid exchanges by inhibitors of topoisomerases. mouse sperm without micronucleus induction in mouse bone marrow
Cell Biol. Toxicol., 2, 485–494. erythrocytes at the same dose level. Mutat. Res., 583, 158–167.
23. Frei, H. and Würgler, F. E. (1996) Induction of somatic mutation and 45. Pacchierotti, F., Ranaldi, R., Eichenlaub-Ritter, U., Attia, S. and
recombination by four inhibitors of eukaryotic topoisomerases assayed in Adler, I. D. (2008) Evaluation of aneugenic effects of bisphenol A in
the wing spot test of Drosophila Melanogaster. Mutagenesis, 11, 315–325. somatic and germ cells of the mouse. Mutat. Res., 651, 64–70.
24. Bakheet, S. A., Attia, S. M., Al-Rasheed, N. M. et al. (2011) Salubrious 46. Adler, I. D. (1993) Synopsis of the in vivo results obtained with the 10
effects of dexrazoxane against teniposide-induced DNA damage and known or suspected aneugens tested in the CEC collaborative study. Mutat.
programmed cell death in murine marrow cells. Mutagenesis, 26, 533–543. Res., 287, 131–137.
25. Attia, S. M., Aleisa, A. M., Bakheet, S. A., Al-Yahya, A. A., Al-Rejaie, S. S., 47. Miller, B. M. and Adler, I. D. (1992) Aneuploidy induction in mouse
Ashour, A. E. and Al-Shabanah, O. A. (2009) Molecular cytogenetic spermatocytes. Mutagenesis, 7, 69–76.
evaluation of the mechanism of micronuclei formation induced by 48. Zucker, R. M., Adams, D. J., Bair, K. W. and Elstein, K. H. (1991)
camptothecin, topotecan and irinotecan. Environ. Mol. Mutagen., 50, Polyploidy induction as a consequence of topoisomerase inhibition. A flow
145–151. cytometric assessment. Biochem. Pharmacol., 42, 2199–2208.

8
Aneugenic effects of teniposide

49. Chen, M. and Beck, W. T. (1995) Differences in inhibition of chromosome


separation and G2 arrest by DNA topoisomerase II inhibitors merbarone
and VM-26. Cancer Res., 55, 1509–1516.
50. Murray, W. A. (1992) Creative blocks: cell-cycle checkpoints and feedback
controls. Nature, 359, 599–604.
51. Morse-Gaudio, M. and Risley, M. S. (1994) Topoisomerase II expression
and VM-26 induction of DNA breaks during spermatogenesis in Xenopus
laevis. J. Cell Sci., 107, 2887–2898.
52. Adler, I. D., Schmid, T. E. and Baumgartner, A. (2002) Induction of
aneuploidy in male mouse germ cells detected by the sperm-FISH assay:
a review of the present data base. Mutat. Res., 504, 173–182.
53. Attia, S. M., Badary, O. A., Hamada, F. M., Hrabé de Angelis, M. and
Adler, I. D. (2008) The chemotherapeutic agents nocodazole and amsacrine
cause meiotic delay and non-disjunction in spermatocytes of mice. Mutat.
Res., 651, 105–113.
54. Baumgartner, A., Czeizel, A. E., Adler, I.-D., Lowe, X., Schmid, T. E. and
Wyrobek, A. J. (1996) Chromosomes X, Y, and 21 aneuploidies in sperm
of men who ingested ultra-high doses of diazepam. Am. J. Hum. Genet., 59
(Suppl. 1), A137.
55. Alberts, B., Bray, D., Lewis, J., Raff, M., Roberts, K. and Watson, J. D.
(1994) Molecular Biology of the Cell. 3rd edn. Garland Publishing, New
York, NY.
56. Haim, N., Roman, J., Nemec, J. and Sinha, B. K. (1986) Peroxidative free
radical formation and O-demethylation of etoposide(VP-16) and teniposi-
de(VM-26). Biochem. Biophys. Res. Commun., 135, 215–220.
57. Attia, S. M. (2010) Deleterious effects of reactive metabolites. Oxid. Med.
Cell Longev., 3, 238–253.
58. Holm, C., Stearns, T. and Botstein, D. (1989) DNA topoisomerase II must

Downloaded from mutage.oxfordjournals.org at King Saud University on September 3, 2011


act at mitosis to prevent nondisjunction and chromosome breakage. Mol.
Cell Biol., 9, 159–168.

View publication stats

You might also like