SlideShare a Scribd company logo
1 of 8
Download to read offline
© The Author 2015. 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.
335
Mutagenesis, 2015, 335–342
doi:10.1093/mutage/gev013
Original Article
Advance Access publication 1 May 2015
Original Manuscript
Integration of Pig-a, micronucleus, chromosome
aberration and comet assay endpoints in a 28-
day rodent toxicity study with urethane
Leon F. Stankowski, Jr1,
*, Marilyn J. Aardema1,2
,Timothy E. Lawlor1
,
Kamala Pant1
, Shambhu Roy1
,Yong Xu1
and Reem Elbekai1
1
BioReliance Corporation, Rockville, MD 20850, USA, 2
Marilyn Aardema Consulting LLC, Fairfield, OH 45014, USA
*To whom correspondence should be addressed. Tel: +1 240 447 2674; Fax: +1 301 738 2362; Email: leon.stankowski@bioreliance.com
Received October 1 2014; Revised January 21 2015; Accepted January 21 2015.
Abstract
As part of the international Pig-a validation trials, we examined the induction of Pig-a mutant
reticulocytes and red blood cells (RETCD59−
and RBCCD59−
, respectively) in peripheral blood of male
Sprague Dawley®
rats treated with urethane (25, 100 and 250 mg/kg/day) or saline by oral gavage
for 29 days. Additional endpoints integrated into this study were: micronucleated reticulocytes
(MN-RET) in peripheral blood; chromosome aberrations (CAb) and DNA damage (%tail intensity
via the comet assay) in peripheral blood lymphocytes (PBL); micronucleated polychromatic
erythrocytes (MN-PCE) in bone marrow; and DNA damage (comet) in various organs at termination
(the 29th dose was added for the comet endpoint at sacrifice). Ethyl methanesulfonate (EMS;
200 mg/kg/day on Days 3, 4, 13, 14, 15, 27, 28 and 29) was evaluated as the concurrent positive
control (PC). All animals survived to termination and none exhibited overt toxicity, but there were
significant differences in body weight and body weight gain in the 250-mg/kg/day urethane group,
as compared with the saline control animals. Statistically significant, dose-dependent increases
were observed for urethane for: RETCD59−
and RBCCD59−
(on Days 15 and 29); MN-RET (on Days 4,
15 and 29); and MN-PCE (on Day 29).The comet assay yielded positive results in PBL (Day 15) and
liver (Day 29), but negative results for PBL (Days 4 and 29) and brain, kidney and lung (Day 29). No
significant increases in PBL CAb were observed at any sample time. Except for PBL CAb (likely due
to excessive cytotoxicity), EMS-induced significant increases in all endpoints/tissues.These results
compare favorably with earlier in vivo observations and demonstrate the utility and sensitivity
of the Pig-a in vivo gene mutation assay, and its ability to be easily integrated, along with other
standard genotoxicity endpoints, into 28-day rodent toxicity studies.
Introduction
The induction and quantification of Pig-a mutant erythrocytes in
rodents were first described in 2008 (1–4). Since then, this assay has
been the subject of intensive study and global validation coordi-
nated by Litron Laboratories and the Genetic Toxicology Technical
Committee of the ILSI Health and Environmental Sciences Institute
(5–16). These studies indicated that the Pig-a endpoint was highly
transferrable and reproducible between laboratories, it could be eas-
ily integrated into standard toxicology studies, and that it held great
promise as an in vivo mutation assay (17,18).
Integration of genotoxicity endpoints into 28-day toxicity studies
is included in new regulatory guidelines and protocols, such as those
from the International Conference on Harmonisation of Technical
Requirements for Registration of Pharmaceuticals for Human Use
[ICH; (19)] and the Organization for Economic Co-operation and
Development [OECD; (20–22)], as a way to maximize genotoxicity
assessments while minimizing the use of animals. As part of these
continuing Pig-a validation trials, we examined the induction of
RETCD59−
and RBCCD59−
in peripheral blood of male Sprague Dawley®
rats treated with urethane for 29 consecutive days. For comparison,
MN-RET in peripheral blood, chromosome aberrations (CAb) and
byguestonJanuary18,2017http://mutage.oxfordjournals.org/Downloadedfrom
DNA damage (via the comet assay) in peripheral blood lymphocytes
(PBL), micronucleated polychromatic erythrocytes (MN-PCE) in
bone marrow and DNA damage in various organs were also evalu-
ated. In this study, we demonstrate how every standard in vivo geno-
toxicity endpoint, including Pig-a, can be integrated into standard
28-day toxicity studies.
Urethane is carcinogenic in multiple rodent species by various
routes (23–25), and we use it at BioReliance as a very effective PC in
ras H2 transgenic carcinogenicity studies (26). It is also mutagenic
and clastogenic in vivo, but generally is considered non-genotoxic
in vitro due to its unique metabolism requirements (24,25,27,29).
Metabolism of urethane requires CYP2E1 and carboxylesterase
isozyme hydrolase A  and proceeds through vinyl carbamate and
conversion to an epoxide that can interact with DNA (29,30).
Support for this metabolic pathway comes from the observation that
urethane-induced MN-PCE frequencies are reduced in CYP2E1-null
mice (31). Thus, urethane represents a key test chemical to evaluate
the sensitivity and applicability of the Pig-a assay, particularly for
the detection of promutagens (18).
Materials and Methods
Reagents
Urethane (CAS no. 51-79-6) and ethyl methanesulfonate (EMS; CAS no.
62-50-0) were purchased from Sigma-Aldrich (St. Louis, MO). Saline
for injection (USP; saline) was purchased from B. Braun Medical (Irvine,
CA). Prototype MutaFlow®
Rat Pig-a Mutation Assay and MicroFlow®
Rat Blood Micronucleus Kits were supplied by Litron Laboratories
(Rochester, NY). Other reagents and supplies included Lympholyte®
-
Mammal cell separation media from Cedarlane (Burlington, NC);
Anti-PE MicroBeads, LS Columns and a QuadroMACS™ Separator
from Miltenyi Biotec (Bergisch Gladbach, Germany); CountBright™
Absolute Count Beads from Invitrogen (Carlsbad, CA).
Animals, treatment and harvests
The number of animals, procedures and design used for this study
were reviewed and approved by the BioReliance Institutional Animal
Care and Use Committee and followed the specifications recom-
mended in the most current version of The Guide for the Care and
Use of Laboratory Animals (32). In addition, the study was conducted
in compliance with Good Laboratory Practice regulations (33).
Five- to six-weeks old Sprague Dawley®
male rats (Harlan; Frederick,
MD) were singly housed in polycarbonate cages and acclimated for
11 days prior to the start of dosing. Certified Rodent Diet (Global Diet
#2018C; Harlan TEKLAD, Madison, WI) and water were provided ad
libitum. Sani-Chip Hardwood bedding (P.J. Murphy Forest Products,
Montville, NJ) was used to absorb liquids. Animals were housed in an
environmentally controlled room with temperatures between 21 and
24°C,a relative humidity of 30–70%,at least 10 changes of fresh HEPA-
filtered air every hour, and a 12-h light/12-h dark cycle. Animals were
7–8-weeks old at the start of dosing and weighed 176–225 g. General
daily clinical observations, as well as weekly detailed observations, were
performed for each animal. Body weight and food consumption were
monitored throughout the treatment period.
Animals were treated for 29 consecutive days with urethane at
doses of 25, 100 and 250 mg/kg/day, or the saline vehicle control (all
n = 10; Table 1). The EMS PC was administered at a dose of 200 mg/
kg/day on Days 3, 4, 13, 14, 15, 27, 28 and 29 (n = 6). The final
dose administered on Day 29 was necessary for the comet endpoint.
Urethane dose formulations were prepared twice (on Days 0 and
14), aliquoted for daily use, stored at 2–8ºC and maintained at room
temperature for at least 30 min prior to use. EMS dose formulations
were freshly prepared and maintained at room temperature until use.
All dose formulations were prepared in saline, corrected for potency
and administered by oral gavage using a 10-ml/kg dose volume, with
the first day of dosing designated as Day 1.
Blood and tissue collection
Blood for the various endpoints was collected from the retro-orbital
sinus under 70% CO2
/30% O2
anesthesia at the volumes and time
points indicated (Table 2). Each collection was ~2.5–3 h after the appro-
priate daily dose as is typical for the comet endpoint (22). Blood for the
comet and the CAb assays was collected directly into sodium heparin
tubes, whereas blood for the Pig-a and MN-RET assays was initially
drawn into an empty tube and then quickly aliquoted into the appropri-
ate volume of heparin (Litron kit supplied). Samples also were collected
for clinical pathology (Clin Path) analysis (hematology, clinical chemis-
try), as is standard for a 28-day rodent toxicity assay (not shown).
All animals were sacrificed by CO2
overdose on Day 29, immedi-
ately after the final blood collections, and a complete necropsy was
performed. The left kidney, and consistent portions of the left lateral
liver lobe, brain, spleen and lungs were collected for the comet assay.
Similarly, the left femur was reserved for bone marrow micronucleus
assessment. All remaining portions of these tissues, as well as numer-
ous other tissues and the entire carcass including animal identifica-
tion, were collected and fixed in 10% neutral buffered formalin for
histopathologic evaluation (as in a standard 28-day rodent toxicity
assay; data not shown).
Pig-a assay
Blood was processed, labeled and analysed using Prototype In
Vivo MutaFlow kits and instructions (version 120209, with minor
Table 1.  Study design
Group Treatment Animals
per groupa
Dose level Dose formulation
(mg/kg/day) (mg/ml)
1 Saline 10 0.00 0.00
2 Urethane 10 25.0 2.50
3 Urethane 10 100 10.0
4 Urethane 10 250 25.0
5 EMS 6 200 20.0
a
Males only. Ten animals per group were treated with urethane or the ve-
hicle control (as per a standard 28-day rodent toxicity study). Eight animals/
group (or all from the EMS positive control group) initially were analysed
prior to dosing to determine the pre-dose Pig-a mutant frequencies. One ani-
mal from the 100 mg/kg/day urethane group was excluded from all subsequent
genetic toxicology endpoint analyses (due to a high spontaneous Pig-a mutant
frequency), and the first six animals remaining in each group were analysed for
all genetic toxicology endpoints.
Table 2.  Sampling times and blood volumes (µl)a
Day N Pig-a MN-RET CAb Comet Clin path Total
−5 or −3 8 100 – – – – 100
4 6 – 150 250 100 – 500
15 6 100 150 250 100 – 600
29 6 100 150 250 100 800 1400
a
Blood was collected ~2.5–3 h after dosing, except for the pre-dose samples
on Days −3 or −5.
336 L. F. Stankowski et al., 2015, Vol. 30, No. 3
byguestonJanuary18,2017http://mutage.oxfordjournals.org/Downloadedfrom
modifications) and with immunomagnetic enrichment (34). A stand-
ard template was used and data was acquired using a BD FACSCanto
II flow cytometer with FACSDiva Software (BD Biosciences; San
Jose, CA). Whenever possible, ≥100 million total RBCs and ≥3 mil-
lion RETs were analysed. For each animal, the total number of RET
and RBC equivalents analysed, as well as the number and frequency
of RETCD59−
and RBCCD59−
, were determined/calculated.
MN-RET assay
Blood was processed, labeled and analysed according to the instruc-
tion manual (version 120702)  received with the MicroFlow®
Rat
Blood Micronucleus Kit. Approximately 20 000 RET were scored
for micronuclei, per animal and time point, where possible. The
same flow cytometer and software were used for data acquisition
and analysis.
MN-PCE assay
Bone marrow was aspirated by syringe, transferred to a centrifuge
tube containing cold fetal bovine serum (FBS), pelleted by centrifu-
gation, resuspended in a small volume of FBS and spread onto a
glass slide (2/animal). The slides were air dried for 24–72 h, fixed in
methanol and stained with acridine orange for microscopic evalua-
tion. Coded slides were scored by fluorescent microscopy at 1000×
magnification for the presence of micronuclei (2000 PCEs/animal;
12 000 per group), and for the proportion of PCEs to total eryth-
rocytes (PCE/EC ratio) as a measure of bone marrow cytotoxicity
(1000 total erythrocytes/animal).
Ex vivo PBL CAb assay
The ex vivo PBL CAb assay was performed and scored as pre-
viously reported (35). Briefly, whole blood was washed by cen-
trifugation using phosphate-buffered saline containing ~2% (v/v)
FBS and resuspended in a 10-fold volume of RPMI 1640 medium
containing 25-mM HEPES, 10% (v/v) heat inactivated FBS, 100-
units/ml penicillin, 100-μg/ml streptomycin, 2-mM L-glutamine
and 20-μg/ml phytohemagglutinin-P to stimulate lymphocyte
proliferation. Single cultures were established in 25-cm2
flasks
(one per animal) and incubated at 37 ± 2°C in a humidified atmos-
phere of ~5% CO2
in air. Cultures were grown for ~48 h, with
Colcemid®
(0.2 μg/ml) being added for the last ~2.5 h before har-
vest. Duplicate slides were prepared from each culture, stained
with Giemsa, air dried and coded, and 100 cells with 2n ± 2 chro-
mosomes were scored per animal for structural aberrations and
numerical aberrations, whereas 500 cells/ animal were evaluated
for mitotic index.
Comet assay
The alkaline comet assay was used to analyze all tissues using the
latest JaCVAM protocol (36,37). Peripheral blood samples were
centrifuged and resuspended in phosphate-buffered saline to pre-
pare comet slides. Liver, kidney, lung, brain and spleen were placed
in cold mincing buffer and finely minced with a pair of fine scis-
sors to release the cells. Each cell suspension was strained through
a 40-µm sieve into a 50-ml-conical tube and stored on wet ice until
the slides were prepared. Approximately 2.5–5 µl of cell suspension
were mixed with 75-µl 0.5% (w/v) low melting agarose and applied
to glass microscope slides (four slides/animal/tissue) that were previ-
ously coated with 1% (w/v) normal melting agarose (to minimize
sample degradation, dosing and necropsy was staggered and highly
choreographed, such that processing up to the application to the
slides was accomplished in ~15 min or less). The slides were kept at
2–8°C for at least 10 to 15 min to solidify.
Following solidification, the slides were submerged in cold lysis
solution (100-mM Na2
EDTA, 2.5-M NaCl, 10-mM Tris, 1% Triton
X-100 and 10% dimethyl sulfoxide; pH 10) for at least overnight
at 2–8°C. After lysis, slides were neutralized in 0.4-M Tris (pH
~7.5), transferred to the electrophoresis chamber which was slowly
filled with cold electrophoresis buffer (300-mM NaOH and 1-mM
Na2
EDTA; pH 13), and maintained for 20 min at 2–10°C, protected
from light, to allow the DNA to unwind. Electrophoresis was con-
ducted for 30 min at 0.7 V/cm, at 2–10°C and protected from light.
After electrophoresis, the slides were removed and washed with neu-
tralization buffer, dehydrated with 200-proof ethanol, air dried and
stored at room temperature with desiccant. Slides were stained with
Sybr-gold™ in Tris-boric acid- EDTA buffer.
Up to 50 randomly selected, nonoverlapping cells were scored
per slide, for a total of 100 cells evaluated per animal and tissue,
using the comet Assay IV scoring system (version 4.11; Perceptive
Instruments Ltd.; Bury St. Edmunds, UK). DNA damage was
assessed based upon %Tail DNA. Each slide was also examined for
indications of possible cytotoxicity (i.e. the percentage of “clouds” in
100 cells/slide, where possible).
Data analyses
Pig-a analyses were performed on log(10) transformed mutant
frequencies (RETCD59−
and RBCCD59−
), and untransformed %RETs,
using the individual animal as the unit of measure. An offset of 0.1
was also added to each mutant RET value prior to transformation
to avoid zero values. One-way analysis of variance was performed
(for mutant frequencies and %RETs), followed by Dunnett’s post
hoc analysis to compare each dose to the vehicle control. A pair-
wise t-test was performed to compare the saline and EMS PCs.
Significance for all statistical analyses was established at the P  0.05
or 0.01 levels using a two-tailed test.
The percent MN-RET and %RET per group were tested for nor-
mal distribution (Levene’s test, P  0.05), and log transformed if not
normally distributed. Urethane data were analysed using one-way
analysis of variance followed by Dunnett’s post hoc analysis. A pair-
wise t-test was performed to compare saline versus the EMS PC.
MN-PCE frequencies were tested for normal distribution
(Levene’s test, P  0.05; and log transformed if not normally distrib-
uted) and analysed using the binomial distribution (Kastenbaum–
Bowman tables). CAb data were analysed using Fisher’s exact test,
while the comet assay results (animal mean calculated from the
median %Tail DNA, 50 cells/slide per animal) were analysed as
described for MN-RET.
Body weight, food consumption and organ weight data were
analysed by Dunnett’s test.
Results
Survival and clinical observations
All animals survived to Day 29 and none exhibited overt signs of
toxicity due to urethane or EMS treatment (data not shown). The
dose level for the EMS PC is the same as we normally use in an
acute, stand-alone comet assay, or in a combination comet/micro-
nucleus study using three daily dose administrations. However, the
cumulative dose and bone marrow toxicity induced by the eight
administrations used here was clearly cytotoxic to the bone mar-
row, as evidenced by the %RET, PCE/EC, and CAb results below.
In contrast, there were no significant changes in Clin Path for the
Integration of Pig-a, micronucleus, chromosome aberration and comet assay endpoints, 2015, Vol. 30, No. 3 337
byguestonJanuary18,2017http://mutage.oxfordjournals.org/Downloadedfrom
urethane-treated animals except for an increase in % monocytes at
a dose of 250 mg/kg/day.
Body weight, body weight change and food
consumption
Significant differences in body weight were noted for the 250-mg/kg/
day urethane treatment group, as well as the EMS PC group, starting
on Day 8 (P  0.05; not shown). In addition, significant differences
in body weight gain were noted during the first 2 weeks of the study,
for the same two groups. Absolute mean body weight gains over the
course of the study, as compared to the control animals, were 22.9%
and 51.4% lower, respectively (not shown). Group mean food con-
sumption values were lower in the 250-mg/kg/day urethane group,
as compared to the control animals, but the differences were not
significant (P  0.05; food consumption data were not collected for
the EMS PC group).
Pig-a assay
Statistically significant, dose-dependent increases in the frequency of
RETCD59−
were observed at doses of 100 and 250 mg/kg/day on Days
15 and 29 (P  0.05; Figure 1, Table 3). The maximum observed
RETCD59−
frequencies were ~74- and 24-fold vehicle control values
on Days 15 and 29, respectively (the relatively smaller increase
on Day 29 was due to the higher mutant frequency in the vehicle
control animals at the later time point). Significant increases in
RBC CD59−
also were observed at a dose of 250 mg/kg/day on Day
15, and at doses of 100 and 250 mg/kg/day on Day 29 (P  0.05;
Figure 2, Table 3). These increases reached maximums of ~4.2- and
8.1-fold control values on Days 15 and 29, respectively.
MN-RET assay
%RET, measured concurrently with the MN-RET endpoint, indi-
cated urethane and the EMS PC reached the bone marrow and
induced significant toxicity. Statistically significant decreases in
%RET were observed for urethane at a dose of 250 mg/kg/day on
Days 4 and 29 (but not Day 15), and for EMS on Days 15 and 29
(Figure 3, Table 3). The apparent lack of bone marrow toxicity for
urethane on Day 15 is curious and may reflect an initial compensa-
tory increase in erythropoiesis.
Statistically significant, dose-dependent increases in the frequency
of MN-RET, to ~4.0- to 6.6-fold control values, were observed
for urethane at all doses on Days 4 and 29, and doses of 100 and
250 mg/kg/day on Day 15 (P  0.05 or 0.01; Figure 4, Table 3). EMS
also induced significant increases in MN-RET at all time points;
these increases were larger at the later two time points, likely due
to the change in dose regimen (two vs. three daily doses before the
scheduled collection times).
MN-PCE assay
Significant, dose-dependent increases in MN-PCE were observed for
urethane-treated animals at terminal sacrifice at a dose of 250 mg/
kg/day (Figure 5; Table 3). EMS induced a much larger increase in
MN-PCE. In contrast to measurements in the peripheral blood, sig-
nificant decreases in PCE/EC ratios were only noted for EMS (not
shown).
Figure 1.  Frequency of RETCD59−
induced by urethane or the EMS PC (all n = 6;
±1 SD). Statistically significant increases are indicated at P  0.05 and 0.01
levels (* and **, respectively). The 95% upper control limit for historical
negative control RETCD59−
data was 2.90 × 10−6
.
Table 3.  Summary of results
Endpoint Maximum fold-increasea
Day 4 Day 15 Day 29
RETCD59−
nd 74 24
RBCCD59−
nd 4.2 8.1
MN-RET 6.6 4.0 5.3
MN-PCE nd nd 5.0
PBL cab – – –
Comet
PBL – 3.0 –
Brain nd nd –
Kidney nd nd –
Liver nd nd 1.5
Lung nd nd –
Spleen nd nd –
nd = not determined.
– = negative response.
a
Only the greatest observed increase, at any dose level or time point, is
reported; any numeric entry represents a statistically significant, positive re-
sponse (P  0.05).
Figure  2.  Frequency of RBCCD59−
induced by urethane or the EMS PC (all
n = 6; ±1 SD). Statistically significant increases are indicated at P  0.05 and
0.01 levels (* and **, respectively).The 95% upper control limit for historical
negative control RBCCD59−
data was 1.39 × 10−6
.
338 L. F. Stankowski et al., 2015, Vol. 30, No. 3
byguestonJanuary18,2017http://mutage.oxfordjournals.org/Downloadedfrom
Ex vivo PBL CAb assay
No significant increases were observed in structural or numerical
aberrations at any dose or sample time for the urethane-treated ani-
mals (results not shown, but summarized in Table 3). This lack of
response was clearly not due to a lack of exposure, since relative
mitotic indices decreased in a dose dependent manner and reached
relative mitotic indices of ~69, 47 and 53% at a dose of 250 mg/kg/
day on Days 4, 15 and 29, respectively. Cultures from EMS-treated
animals were unable to be scored due to insufficient metaphases,
likely as the result of excessive toxicity (relative mitotic indices were
~17, 10 and 9% of control values on Days 4, 15 and 29, respectively).
Comet assay
Statistically significant, dose-dependent increases in median %Tail
DNA were observed for urethane only in PBL at Day 15, and in liver
at terminal sacrifice (P  0.05; Figures 6 and 7; Table 3). Urethane
was considered to be negative in all other tissues and time points
analysed.The EMS PC induced significant increases in median %Tail
DNA in all tissues and at all sampling times (P  0.05). The percent-
age of clouds observed in the urethane-treated animals was compa-
rable to, or less than, that observed in the vehicle control animals,
for all tissues and time points (not shown). In contrast, increases in
the percentage of clouds in the EMS-treated animals ranged from
~2- to 4-fold (blood on Day 4 and brain, lung and spleen) to ~8-fold
or greater in the other tissues, and blood at the later time points (not
shown).
Discussion
One goal of this study was to validate the integration of the in
vivo Pig-a assay, as well as other genetic toxicology endpoints, into
a typical 28-day rodent toxicity assay in compliance with Good
Laboratory Practice regulations (33). As such, all of the typical body
and organ weight, cage-side and detailed observations, food con-
sumption and Clin Path procedures/analyses were performed (tissues
were collected for histopathology but not analysed due to the time
and expense involved). Only the results relevant to interpretation of
the effects of urethane are reported here.
Based on data in the literature, urethane was positive in most in
vivo cytogenetic and mutation studies in vivo (24,25,28). Results
from our study compare favorably with these previous in vivo obser-
vations as discussed below. Urethane induced statistically significant,
dose-dependent increases in the frequencies of MN-RET at all time
points analysed, and in MN-PCE at terminal sacrifice. The maximal
fold-increases observed were similar (~4- to 6-fold for all observa-
tions), even though the absolute frequencies were quite different for
MN-RET and MN-PCE. It should be noted that positive (significant)
responses were observed for MN-RET at doses ≥100 mg/kg/day, but
only at 250 mg/kg/day for the MN-PCE endpoint. This difference
is likely due to the improved sensitivity and power afforded by the
ability to routinely score many more cells using the automated flow
cytometric method (38). Also, the apparent “disappearance” of the
positive MN-RET response at a dose of 25.0 mg/kg/day on Day 15
may reflect that the dose/response was at or near the limit of detec-
tion for the assay.
Urethane also induced statistically significant, dose-dependent
increases in the comet assay (median %Tail DNA) in PBL on Day
15, and in liver at terminal sacrifice. However, the relative increases
(1.5- to 3.0-fold control values) were even less than observed for
MN-RET and MN-PCE. In addition, no significant increases were
observed for the comet endpoint in blood on Day 4 or 29. Thus, the
Figure 3.  Percent RET in peripheral blood, following treatment with urethane
or the EMS PC, as assessed by flow cytometry (all n = 6; ±1 SD).
Figure  4.  Frequency of MN-RET induced by urethane or the EMS PC (all
n = 6; ±1 SD). Statistically significant increases are indicated at P  0.05 and
0.01 levels (* and **, respectively).The 95% upper control limit for historical
negative control MN-RET data was 0.175%.
Figure  5.  Frequency of MN-PCE induced by urethane or the EMS PC (all
n = 6; ±1 SD). Statistically significant increases are indicated at P  0.05 and
0.01 levels (* and **, respectively).The 95% upper control limit for historical
negative control MN-PCE data was 0.0947%.
Integration of Pig-a, micronucleus, chromosome aberration and comet assay endpoints, 2015, Vol. 30, No. 3 339
byguestonJanuary18,2017http://mutage.oxfordjournals.org/Downloadedfrom
“positive” comet response, especially in blood, is considered to be
suspect. An apparent dose-dependent increase in %Tail DNA was
observed in lung, but the individual values did not reach statisti-
cal significance (P  0.05). Although dose-dependent decreases in
median %Tail DNA were observed in the brain, the assay condi-
tions were not optimized for detection of crosslinkers nor was that
intended for this study. The lack of any appreciable increases in
clouds and the lack of any significant deleterious changes in Clin
Path parameters suggest that any significant increases in comet
response were not due to cytotoxicity.
Also to our knowledge, this study represents the first reported
in vivo comet assay with urethane. The negative comet results in
lung are perhaps unexpected, since it is one of the principal sites for
tumor formation (24,25), and positive results have been observed in
transgenic rodent mutation (TRM) assays, in multiple tissues includ-
ing forestomach, liver and lung, following oral dosing [reviewed in
refs. (39–41)]. However, in the cases of earlier positive TRM results,
the cumulative dose levels generally were much higher, and/or the
sampling time was much later, than studied here (see additional
discussion below regarding cumulative dose and sampling times).
Likewise, the sampling time was our standard, in compliance with
OECD recommendations (22), but not necessarily optimal based
upon uptake and metabolism kinetics or DNA repair.
No significant increases were observed for urethane in the CAb
assay at any dose or sampling time. In searching the literature, we
found no reports of structural CAb induced by urethane when
scored by metaphase analysis; rather, the induction of CAb was
inferred using micronuclei as a surrogate for chromosome breakage.
Although the micronucleus test should be considered the in vivo test
of choice for CAb (21), little additional effort was required to add
the CAb endpoint to this study, as has been suggested elsewhere (42).
Since a daily dosing regimen was used, and the 48-h culture time
after stimulation has proved reliable in this laboratory, the recom-
mended sampling time for the comet assay that was used here (22)
is likely not responsible for the lack of a positive CAb response for
urethane. However, we also note the PCs for this assay failed, but
likely due to excessive toxicity.
Finally, and most importantly for this study, urethane induced
statistically significant, dose-dependent increases in the frequen-
cies of RETCD59−
and RBCCD59−
at both time points analysed. Larger
responses were observed in RETCD59−
, which is consistent with earlier
observations for other agents and is due to slower turnover in the
RBC population (18). The maximum mutant frequencies observed
at a dose of 250 mg/kg/day were ~20 × 10−6
for RETCD59−
on Day 15,
and ~6 × 10−6
for RBCCD59−
on Day 29. These numbers compare very
favorably to those observed for urethane used under identical con-
ditions as a PC in a study of methyl carbamate (43). The relatively
larger fold-increase in RETCD59−
on Day 15 versus Day 29 (Table 3)
is the result of an approximate 3-fold difference in negative con-
trol mutant frequencies at those time points (2.82 vs. 0.678 × 10−6
,
respectively).
The 24- and 74-fold increases in RETCD59−
frequencies on Day 29
and 15 far exceeded those observed in the other positive endpoints.
Based upon the biology and expression time for the RETCD59−
and
RBCCD59−
to appear in the peripheral blood, and as seen for a num-
ber of chemicals, at least some additional increases in mutant fre-
quencies could be expected (5,6,18,44,45). Given sufficient time, it
is anticipated that the fold-increases in RBCCD59−
frequencies would
reach levels similar to those observed for RETCD59−
. These results,
in comparison to the other endpoints studied, further reinforce the
dramatic benefits of repeat dosing, and accumulation of induced
mutants, that are associated with the Pig-a assay [as has been
reported previously and reviewed by Gollapudi et al. (18)].
Urethane has been reported to be one of the weakest mutagens
examined to date in the TRM assays using the currently recom-
mended study design (46–48). Only half of the urethane treatments
previously reviewed (28/56) produced positive findings [by any
route; results compiled in refs. (39–41)]. Although mutant frequen-
cies in the lung and liver reached ~5- to 19-fold control values in
some of these TRM studies (representing induced mutant frequen-
cies of ~800–1300 × 10‒6
), these were 6- to 8-month drinking water
or feed studies performed over much longer intervals (105–237 days)
at ~2- to 10-fold higher cumulative doses (13 650–69 678 mg/kg)
than in our study (7250 mg/kg). Most other oral dosing TRM studies
(gavage or diet) reported in the literature using dose regimens similar
to our study (1400–5600 mg/kg by oral gavage) resulted in approxi-
mately only 2-fold increases in mutant frequencies in the lung or
liver (representing induced mutant frequencies of ~60–110 × 10−6
)
or negative results (39–41). Similar TRM studies (cumulative doses
Figure  7.  Comet analysis in various tissues following 29 daily doses of
urethane (median %Tail DNA ±1 SD; n  =  6, except as noted). EMS PC
responses were off scale (~30–34%). Statistically significant increase
indicated at P  0.05 level (*). Lung results from one animal were excluded
as an extreme outlier (13.1%). The 95% upper control limits for historical
negative control median %Tail DNA data were: 2.48% (liver), 22.3% (brain),
4.20% (spleen), 4.14% (kidney) and 11.6% (lung).
Figure 6.  Comet analysis in PBL following 29 daily doses of urethane (median
%Tail DNA; all n = 6; ±1 SD). EMS PC responses were off scale (~16–26%).
Statistically significant increases are indicated at P  0.01 level (**).The 95%
upper control limit for historical negative control median %Tail DNA data
was 0.750%.
340 L. F. Stankowski et al., 2015, Vol. 30, No. 3
byguestonJanuary18,2017http://mutage.oxfordjournals.org/Downloadedfrom
of 700 to 5600 mg/kg by oral gavage) produced ~1.7- to 2.8-fold
increases in mutant frequencies in haematopoietic tissues (represent-
ing induced mutant frequencies of ~35–100 × 10−6
). Although the
net increases in Pig-a mutant frequencies reported here were smaller
than some observed in the TRM assays, the fold-increases were
larger due to the lower spontaneous mutant frequencies for Pig-a.
Also as previously stated, the Pig-a mutant frequencies would be
expected to increase with a longer phenotypic expression time.
These results demonstrate the utility and sensitivity of the Pig-a
in vivo gene mutation assay. This endpoint, as well as the other
genetic toxicology endpoints, can be integrated into repeat dose tox-
icity studies easily and economically, resulting in a comprehensive
analysis of genotoxicity with reduced animal use. The relative sensi-
tivity of the various endpoints is likely due in part to the statistical
power and sample sizes of the various assays, and the ability of Pig-a
mutant cells to persist and accumulate during repeated dosing.
The negative and/or weak CAb and comet responses are simi-
lar to our previous observations for 4-nitroquinoline oxide using a
29-day dose regimen (16). Those results, and the weak MN-PCE and
comet responses here, further demonstrate that not all genotoxicity
endpoints respond similarly. Although it is certainly possible to inte-
grate multiple (or all of these) endpoints into a single study, it is not
necessarily prudent or scientifically justified. As previously discussed
(45), the Pig-a and MN-RET are easily integrated, complimentary
endpoints that offer the ability to detect gene mutation and cytoge-
netic damage, sometimes arising from two distinct modes of action,
that cannot be universally detected for all agents by a single assay.
Where limitations exist (e.g. questions over target tissue exposure),
then the comet assay would be a valuable addition. Thus, care must
be taken in dose, endpoint and tissue selection when incorporating
genotoxicity endpoints into routine toxicity studies as has been rec-
ommended, for example, by the ICH (19), the OECD (20–22) and
the US Food and Drug Administration (42).
Funding
This work was supported by BioReliance Corporation and also sup-
ported in part by a grant to Litron Laboratories (National Institute
of Health/National Institute of Environmental Health Sciences,
R44ES018017).
Acknowledgements
The authors thank Sarah Miller for her expert technical assistance, and
Stephen Dertinger for invaluable advice and support. The contents herein are
solely the responsibility of the authors, and do not necessarily represent the
official views of the institutions involved.
Conflict of interest statement: All of the authors are employees of
BioReliance Corporation and M.J.A. also serves as a consultant to
BioReliance. BioReliance is a contract research organization offering
these and other studies for sale.
References
	1.	 Bryce, S. M., Bemis, J. C. and Dertinger, S. D. (2008) In vivo mutation
assay based on the endogenous Pig-a locus. Environ. Mol. Mutagen., 49,
256–264.
	2.	 Miura, D., Dobrovolsky, V. N., Kasahara, Y., Katsuura, Y. and Heflich,
R. H. (2008) Development of an in vivo gene mutation assay using the
endogenous Pig-A gene: I. Flow cytometric detection of CD59-negative
peripheral red blood cells and CD48-negative spleen T-cells from the rat.
Environ. Mol. Mutagen., 49, 614–621.
	3.	 Miura, D., Dobrovolsky, V. N., Mittelstaedt, R. A., Kasahara, Y., Katsuura,
Y. and Heflich, R. H. (2008) Development of an in vivo gene mutation
assay using the endogenous Pig-A gene: II. Selection of Pig-A mutant rat
spleen T-cells with proaerolysin and sequencing Pig-A cDNA from the
mutants. Environ. Mol. Mutagen., 49, 622–630.
	4.	 Phonethepswath, S., Bryce, S. M., Bemis, J. C. and Dertinger, S. D. (2008)
Erythrocyte-based Pig-a gene mutation assay: demonstration of cross-spe-
cies potential. Mutat. Res., 657, 122–126.
	5.	 Phonethepswath, S., Franklin, D., Torous, D. K. et al. (2010) Pig-a muta-
tion: kinetics in rat erythrocytes following exposure to five prototypical
mutagens. Toxicol. Sci., 114, 59–70.
	6.	 Dertinger, S. D., Phonethepswath, S., Franklin, D. et al. (2010) Integration
of mutation and chromosomal damage endpoints into 28-day repeat dose
toxicology studies. Toxicol. Sci., 115, 401–411.
	7.	 Bhalli, J. A., Shaddock, J. G., Pearce, M. G., Dobrovolsky, V. N., Cao, X.,
Heflich, R. H. and Vohr, H. W. (2011) Report on stage III Pig-a mutation
assays using benzo[a]pyrene. Environ. Mol. Mutagen., 52, 731–737.
	8.	 Bhalli, J. A., Pearce, M. G., Dobrovolsky, V. N. and Heflich, R. H. (2011)
Manifestation and persistence of Pig-a mutant red blood cells in C57BL/6
mice following single and split doses of N-ethyl-N-nitrosourea. Environ.
Mol. Mutagen., 52, 766–773.
	9.	 Cammerer, Z., Bhalli, J. A., Cao, X. et al. (2011) Report on stage III Pig-a
mutation assays using N-ethyl-N-nitrosourea-comparison with other in
vivo genotoxicity endpoints. Environ. Mol. Mutagen., 52, 721–730.
	10.	Dertinger, S. D., Phonethepswath, S., Weller, P. et al. (2011) International
Pig-a gene mutation assay trial: evaluation of transferability across 14
laboratories. Environ. Mol. Mutagen., 52, 690–698.
	11.	Dertinger, S. D., Phonethepswath, S., Weller, P. et al. (2011) Interlaboratory
Pig-a gene mutation assay trial: studies of 1,3-propane sultone with immu-
nomagnetic enrichment of mutant erythrocytes. Environ. Mol. Mutagen.,
52, 748–755.
	12.	Kimoto, T., Chikura, S., Suzuki, K. et al. (2011) Further development of
the rat Pig-a mutation assay: measuring rat Pig-a mutant bone marrow
erythroids and a high throughput assay for mutant peripheral blood retic-
ulocytes. Environ. Mol. Mutagen., 52, 774–783.
	13.	Lemieux, C. L., Douglas, G. R., Gingerich, J., Phonethepswath, S., Torous,
D. K., Dertinger, S. D., Phillips, D. H., Arlt, V. M. and White, P. A. (2011)
Simultaneous measurement of benzo[a]pyrene-induced Pig-a and lacZ
mutations, micronuclei and DNA adducts in Muta™ Mouse. Environ.
Mol. Mutagen., 52, 756–765.
	14.	Lynch, A. M., Giddings, A., Custer, L., Gleason, C., Henwood, A., Aylott, M.
and Kenny, J. (2011) International Pig-a gene mutation assay trial (stage III):
results with N-methyl-N-nitrosourea. Environ. Mol. Mutagen., 52, 699–710.
	15.	Shi, J., Krsmanovic, L., Bruce, S. et al. (2011) Assessment of genotoxicity
induced by 7,12-dimethylbenz(a)anthracene or diethylnitrosamine in the
Pig-a, micronucleus and Comet assays integrated into 28-day repeat dose
studies. Environ. Mol. Mutagen., 52, 711–720.
	16.	Stankowski, L. F. Jr, Roberts, D. J., Chen, H., Lawlor, T., McKeon, M.,
Murli, H., Thakur, A. and Xu, Y. (2011) Integration of Pig-a, micronu-
cleus, chromosome aberration, and Comet assay endpoints in a 28-day
rodent toxicity study with 4-nitroquinoline-1-oxide. Environ. Mol. Muta-
gen., 52, 738–747.
	17.	Schuler, M., Gollapudi, B. B., Thybaud, V. and Kim, J. H. (2011) Need
and potential value of the Pig-ain vivo mutation assay-a HESI perspective.
Environ. Mol. Mutagen., 52, 685–689.
	18.	Gollapudi, B.B., Lynch, A.M., Heflich, R.H. et  al. (2014) The in vivo
Pig-a mutant assay: a report of the International Workshop on Geno-
toxicity Testing (IWGT) workgroup. Mutat. Res. doi:10.1016/j.mrgen-
tox.2014.09.007.
	19.	International Conference on Harmonization. (2008) Guidance on geno-
toxicity testing and data interpretation for pharmaceuticals intended for
human use S2(R1) (09 November 2011). Available at: http://www.ich.org/
fileadmin/Public_Web_Site/ICH_Products/Guidelines/Safety/S2_R1/Step4/
S2R1_Step4.pdf (accessed 21 January 2015).
	20.	Organisation of Economic Co-operation and Development. (2014) Guide-
line for Testing of Chemicals: Test Guideline 474 - Mammalian Erythro-
cyte Micronucleus Test, adopted 26 September 2014, OECD, Paris. Avail-
Integration of Pig-a, micronucleus, chromosome aberration and comet assay endpoints, 2015, Vol. 30, No. 3 341
byguestonJanuary18,2017http://mutage.oxfordjournals.org/Downloadedfrom
able at: http://www.oecd-ilibrary.org/docserver/download/9714541e.pdf
(accessed 21 January 2015).
	21.	Organisation of Economic Co-operation and Development. (2014)
Guideline for Testing of Chemicals: Test Guideline 475 - Mammalian
Bone Marrow Chromosomal Aberration Test, adopted 26 September
2014, OECD, Paris. Available at: http://www.oecd-ilibrary.org/docserver/
download/9714551e.pdf (accessed 21 January 2015).
	22.	Organisation of Economic Co-operation and Development. (2014) Guide-
line for Testing of Chemicals: Test Guideline 489 - In Vivo Mammalian
Alkaline Comet Assay, adopted 26 September 2014, OECD, Paris. Avail-
able at: http://www.oecd-ilibrary.org/docserver/download/9714511e.pdf
(accessed 21 January 2015).
	23.	Salmon, A.G. and Zeise, L. (1991) Risks of Carcinogenesis from Urethane
Exposure. CRC Press, Boca Raton, FL.
	24.	International Agency for Research on Cancer. (2010). Ethyl carbamate
(urethane). IARC Monogr. Carcinog, Risks, Hum, 96, 1281–1307.
	25.	European Commission. (2012) Recommendation from the Scientific Com-
mittee on Occupational Exposure Limits for Ethyl Carbamate [Urethane].
SCOEL/SUM/172. Available at http://ec.europa.eu/social/BlobServlet?doc
Id=7723langId=en (accessed 21 January 2015).
	26.	Shah, S. A., Paranjpe, M. G., Atkins, P. I. and Zahalka, E. A. (2012) Reduc-
tion in the number of animals and the evaluation period for the positive
control group in Tg.rasH2 short-term carcinogenicity studies. Int. J. Toxi-
col., 31, 423–429.
	27.	Sotomayor, R. E. and Collins, T. F. (1990) Mutagenicity, metabolism, and
DNA interactions of urethane. Toxicol. Ind. Health, 6, 71–108.
	28.	Tweats, D.J., Blakey, D., Heflich, R.H. et al. (2007) Report of the IWGT
working group on strategy/interpretation of regulatory in vivo tests II.
Identification of in vivo-only positive compounds in the bone marrow
micronucleus test. Mutat. Res. 627, 92–105.
	29.	Forkert, P. G. and Lee, R. P. (1997) Metabolism of ethyl carbamate by
pulmonary cytochrome P450 and carboxylesterase isozymes: involvement
of CYP2E1 and hydrolase A. Toxicol. Appl. Pharmacol., 146, 245–254.
	30.	Guengerich, F. P. and Kim, D. H. (1991) Enzymatic oxidation of ethyl car-
bamate to vinyl carbamate and its role as an intermediate in the formation
of 1,N6-ethenoadenosine. Chem. Res. Toxicol., 4, 413–421.
	31.	Hoffler, U., Dixon, D., Peddada, S. and Ghanayem, B. I. (2005) Inhibition
of urethane-induced genotoxicity and cell proliferation in CYP2E1-null
mice. Mutat. Res., 572, 58–72.
	32.	National Research Council, Institute of Laboratory Animal Resources
Commission on Life Sciences. (2011) Guide for the Care and Use of Labo-
ratory Animals. National Academy Press, Washington, DC.
	33.	United States Food and Drug Administration. (1987) Good Laboratory
Practice for Nonclinical Laboratory Studies. Code of Federal Regulations,
Title 21, Part 58. Federal Register, 52, 33768–33782.
	34.	Dertinger, S. D., Bryce, S. M., Phonethepswath, S. and Avlasevich, S. L.
(2011) When pigs fly: immunomagnetic separation facilitates rapid deter-
mination of Pig-a mutant frequency by flow cytometric analysis. Mutat.
Res., 721, 163–170.
	35.	Erexson, G.L., Farabaugh, C.S., Yung, K.M., and Stojhovic, G.P. 2004. An
in vivo-in vitro study protocol for the conduct of the rat peripheral blood
lymphocyte (RPBL) chromosome aberrations (CA) assay. Environ. Mol.
Mutagen., 44, 198.
	36.	Validation Management Team (VMT) coordinated by JaCVAM (Japanese
Committee for the Validation of the Alternative Methods). (2009) Inter-
national Validation of the In Vivo Rodent Alkaline Comet Assay for the
Detection of Genotoxic Carcinogens (Protocol Version 14.2.). Available at
http://www.oecd.org/officialdocuments/publicdisplaydocumentpdf/?cote=
ENV/JM/MONO(2014)10doclanguage=en (accessed 21 January 2015).
	37.	Rothfuss, A., O’Donovan, M., De Boeck, M. et al. (2010) Collaborative
study on fifteen compounds in the rat-liver Comet assay integrated into
2- and 4-week repeat-dose studies. Mutat. Res., 702, 40–69.
	38.	Kissling, G. E., Dertinger, S. D., Hayashi, M. and MacGregor, J. T.
(2007) Sensitivity of the erythrocyte micronucleus assay: dependence
on number of cells scored and inter-animal variability. Mutat. Res., 634,
235–240.
	39.	Organisation for Economic Cooperation and Development. (2009)
Detailed Review Paper on Transgenic Rodent Mutation Assays, Series on
Testing and Assessment, N° 103. ENV/JM/MONO(2009)7. OECD, Paris.
Available at: http://www.oecd.org/officialdocuments/publicdisplaydocume
ntpdf/?cote=env/jm/mono(2009)7doclanguage=en (accessed 21 January
2015).
	40.	Organisation for Economic Cooperation and Development. (2009)
Part 2: Annexes to the Detailed Review Paper on Transgenic Rodent
Mutation Assays, Series on Testing and Assessment, N° 103. ENV/
JM/MONO(2009)29. OECD, Paris. Available at: http://www.oecd.
org/officialdocuments/publicdisplaydocumentpdf/?cote=env/jm/
mono(2009)29doclanguage=en (accessed 21 January 2015).
	41.	Organisation for Economic Cooperation and Development. (2011) Trans-
genic Rodent Somatic and Germ Cell Gene Mutation Assay: Retrospective
Performance Assessment, Series on Testing and Assessment, N° 145. ENV/
JM/MONO(2011)20. OECD, Paris. Available at: http://www.oecd.org/
env/ehs/testing/48532170.pdf (accessed 21 January 2015).
	42.	United States Food and Drug Administration. (2006) Guidance for indus-
try and review staff: recommended approaches to integration of genetic
toxicology study results. Available at: www.fda.gov/downloads/Drugs/
GuidanceComplianceRegulatoryInformation/Guidances/ucm079257.pdf
(accessed 21 January 2015).
	43.	Bemis, J.C, Labash, C., Avlasevich, S.L., Carlson, K., Berg, A., Torous, D.K.,
Barragato, M., MacGregor, J.T., and Dertinger, S.D. (2015) Rat Pig-a muta-
tion assay responds to the genotoxic carcinogen ethyl carbamate but not
the non-genotoxic carcinogen methyl carbamate. Mutagen. 30, 343–347.
doi:10.1093/mutage/geu084.
	44.	Miura, D., Dobrovolsky, V. N., Kimoto, T., Kasahara, Y. and Heflich,
R. H. (2009) Accumulation and persistence of Pig-A mutant peripheral
red blood cells following treatment of rats with single and split doses of
N-ethyl-N-nitrosourea. Mutat. Res., 677, 86–92.
	45.	Dertinger, S. D., Phonethepswath, S., Avlasevich, S. L. et al. (2012) Effi-
cient monitoring of in vivo pig-a gene mutation and chromosomal dam-
age: summary of 7 published studies and results from 11 new reference
compounds. Toxicol. Sci., 130, 328–348.
	46.	Heddle, J. A., Dean, S., Nohmi, T. et al. (2000) In vivo transgenic mutation
assays. Environ. Mol. Mutagen., 35, 253–259.
	47.	Thybaud, V., Dean, S., Nohmi, T. et al. (2003) In vivo transgenic mutation
assays. Mutat. Res., 540, 141–151.
	48.	Organisation of Economic Co-operation and Development. (2013)
Guideline for Testing of Chemicals: Test Guideline 488 - Transgenic
Rodent Somatic and Germ Cell Gene Mutation Assays, adopted 26 July
2013, OECD, Paris. Available at: http://www.oecd-ilibrary.org/docserver/
download/9713251e.pdf (accessed 21 January 2015).
342 L. F. Stankowski et al., 2015, Vol. 30, No. 3
byguestonJanuary18,2017http://mutage.oxfordjournals.org/Downloadedfrom

More Related Content

What's hot

CYP2A6_HPLC_PK_2015 New Simple Method for Coumarin in Liver Cytochrome of Rats
CYP2A6_HPLC_PK_2015 New Simple Method for Coumarin in Liver Cytochrome of RatsCYP2A6_HPLC_PK_2015 New Simple Method for Coumarin in Liver Cytochrome of Rats
CYP2A6_HPLC_PK_2015 New Simple Method for Coumarin in Liver Cytochrome of RatsWael Ebied
 
Evaluation of Metabolic Stability of Kinsenoside, an Antidiabetic Candidate, ...
Evaluation of Metabolic Stability of Kinsenoside, an Antidiabetic Candidate, ...Evaluation of Metabolic Stability of Kinsenoside, an Antidiabetic Candidate, ...
Evaluation of Metabolic Stability of Kinsenoside, an Antidiabetic Candidate, ...Cây thuốc Việt
 
Impact_of_di-_2-ethylhexyl_phthalate_on
Impact_of_di-_2-ethylhexyl_phthalate_onImpact_of_di-_2-ethylhexyl_phthalate_on
Impact_of_di-_2-ethylhexyl_phthalate_onKalaivani Manokaran
 
Springer_CurrentMicrobiol_Mukherjee_2006
Springer_CurrentMicrobiol_Mukherjee_2006Springer_CurrentMicrobiol_Mukherjee_2006
Springer_CurrentMicrobiol_Mukherjee_2006Dr.Gunjan Mukherjee
 
1 s2.0-s105046481300034 x-main
1 s2.0-s105046481300034 x-main1 s2.0-s105046481300034 x-main
1 s2.0-s105046481300034 x-mainalem010
 
special project cyp2e1 report
special project cyp2e1 reportspecial project cyp2e1 report
special project cyp2e1 reportKemal Asik
 
Effect of astaxanthin on ethylene glycol induced nephrolithiasis
Effect of astaxanthin on ethylene glycol induced nephrolithiasisEffect of astaxanthin on ethylene glycol induced nephrolithiasis
Effect of astaxanthin on ethylene glycol induced nephrolithiasisIOSR Journals
 
The activities of pumelo fruit juice (citrus maxima var
The activities of pumelo fruit juice (citrus maxima varThe activities of pumelo fruit juice (citrus maxima var
The activities of pumelo fruit juice (citrus maxima varAlexander Decker
 
Effect of mow procedure on physiological and biochemical properties of blood ...
Effect of mow procedure on physiological and biochemical properties of blood ...Effect of mow procedure on physiological and biochemical properties of blood ...
Effect of mow procedure on physiological and biochemical properties of blood ...iosrjce
 

What's hot (19)

CYP2A6_HPLC_PK_2015 New Simple Method for Coumarin in Liver Cytochrome of Rats
CYP2A6_HPLC_PK_2015 New Simple Method for Coumarin in Liver Cytochrome of RatsCYP2A6_HPLC_PK_2015 New Simple Method for Coumarin in Liver Cytochrome of Rats
CYP2A6_HPLC_PK_2015 New Simple Method for Coumarin in Liver Cytochrome of Rats
 
Evaluation of Metabolic Stability of Kinsenoside, an Antidiabetic Candidate, ...
Evaluation of Metabolic Stability of Kinsenoside, an Antidiabetic Candidate, ...Evaluation of Metabolic Stability of Kinsenoside, an Antidiabetic Candidate, ...
Evaluation of Metabolic Stability of Kinsenoside, an Antidiabetic Candidate, ...
 
E45012938
E45012938E45012938
E45012938
 
articulo
articulo articulo
articulo
 
Impact_of_di-_2-ethylhexyl_phthalate_on
Impact_of_di-_2-ethylhexyl_phthalate_onImpact_of_di-_2-ethylhexyl_phthalate_on
Impact_of_di-_2-ethylhexyl_phthalate_on
 
Springer_CurrentMicrobiol_Mukherjee_2006
Springer_CurrentMicrobiol_Mukherjee_2006Springer_CurrentMicrobiol_Mukherjee_2006
Springer_CurrentMicrobiol_Mukherjee_2006
 
B0343014018
B0343014018B0343014018
B0343014018
 
Prophylactic Effects of Losartan in Intestinal Ischemia-Reperfusion Injury Model
Prophylactic Effects of Losartan in Intestinal Ischemia-Reperfusion Injury ModelProphylactic Effects of Losartan in Intestinal Ischemia-Reperfusion Injury Model
Prophylactic Effects of Losartan in Intestinal Ischemia-Reperfusion Injury Model
 
Haseeb ahsan
Haseeb ahsanHaseeb ahsan
Haseeb ahsan
 
Article wjpps 1454479295 (2)
Article wjpps 1454479295 (2)Article wjpps 1454479295 (2)
Article wjpps 1454479295 (2)
 
1 s2.0-s105046481300034 x-main
1 s2.0-s105046481300034 x-main1 s2.0-s105046481300034 x-main
1 s2.0-s105046481300034 x-main
 
BBRC pcsk9
BBRC pcsk9BBRC pcsk9
BBRC pcsk9
 
special project cyp2e1 report
special project cyp2e1 reportspecial project cyp2e1 report
special project cyp2e1 report
 
Mon article
Mon articleMon article
Mon article
 
28 hari
28 hari28 hari
28 hari
 
Matsunami et al., 2010 4th
Matsunami et al., 2010 4thMatsunami et al., 2010 4th
Matsunami et al., 2010 4th
 
Effect of astaxanthin on ethylene glycol induced nephrolithiasis
Effect of astaxanthin on ethylene glycol induced nephrolithiasisEffect of astaxanthin on ethylene glycol induced nephrolithiasis
Effect of astaxanthin on ethylene glycol induced nephrolithiasis
 
The activities of pumelo fruit juice (citrus maxima var
The activities of pumelo fruit juice (citrus maxima varThe activities of pumelo fruit juice (citrus maxima var
The activities of pumelo fruit juice (citrus maxima var
 
Effect of mow procedure on physiological and biochemical properties of blood ...
Effect of mow procedure on physiological and biochemical properties of blood ...Effect of mow procedure on physiological and biochemical properties of blood ...
Effect of mow procedure on physiological and biochemical properties of blood ...
 

Viewers also liked

FLC eLearning July2015
FLC eLearning July2015FLC eLearning July2015
FLC eLearning July2015Kwok Tse
 
2.2. el uso especializado de las palabras
2.2. el uso especializado de las palabras2.2. el uso especializado de las palabras
2.2. el uso especializado de las palabrasbibloranca
 
Thiết kế dây chuyền may bài 3 tính toán công nghệ
Thiết kế dây chuyền may   bài 3 tính toán công nghệThiết kế dây chuyền may   bài 3 tính toán công nghệ
Thiết kế dây chuyền may bài 3 tính toán công nghệTÀI LIỆU NGÀNH MAY
 
WEF_AM17_FutureofRetailInsightReport
WEF_AM17_FutureofRetailInsightReportWEF_AM17_FutureofRetailInsightReport
WEF_AM17_FutureofRetailInsightReportSarah Shellaby
 

Viewers also liked (10)

learning styles
learning styleslearning styles
learning styles
 
Les crues torrentielles
Les crues torrentiellesLes crues torrentielles
Les crues torrentielles
 
Suresh bangaru 2016
Suresh bangaru 2016Suresh bangaru 2016
Suresh bangaru 2016
 
Presentation Project
Presentation ProjectPresentation Project
Presentation Project
 
FLC eLearning July2015
FLC eLearning July2015FLC eLearning July2015
FLC eLearning July2015
 
Tipos de virus
Tipos de virusTipos de virus
Tipos de virus
 
2.2. el uso especializado de las palabras
2.2. el uso especializado de las palabras2.2. el uso especializado de las palabras
2.2. el uso especializado de las palabras
 
Thiết kế dây chuyền may bài 3 tính toán công nghệ
Thiết kế dây chuyền may   bài 3 tính toán công nghệThiết kế dây chuyền may   bài 3 tính toán công nghệ
Thiết kế dây chuyền may bài 3 tính toán công nghệ
 
WEF_AM17_FutureofRetailInsightReport
WEF_AM17_FutureofRetailInsightReportWEF_AM17_FutureofRetailInsightReport
WEF_AM17_FutureofRetailInsightReport
 
ni_p_17-01_
ni_p_17-01_ni_p_17-01_
ni_p_17-01_
 

Similar to Pig-a

Jianying Xiao CV 2016
Jianying Xiao CV 2016  Jianying Xiao CV 2016
Jianying Xiao CV 2016 Xiao Jianying
 
IOSR Journal of Pharmacy (IOSRPHR), www.iosrphr.org, call for paper, research...
IOSR Journal of Pharmacy (IOSRPHR), www.iosrphr.org, call for paper, research...IOSR Journal of Pharmacy (IOSRPHR), www.iosrphr.org, call for paper, research...
IOSR Journal of Pharmacy (IOSRPHR), www.iosrphr.org, call for paper, research...iosrphr_editor
 
Medicilon DMPK & Bioanalysis Services-Preclinical Research
Medicilon DMPK & Bioanalysis Services-Preclinical ResearchMedicilon DMPK & Bioanalysis Services-Preclinical Research
Medicilon DMPK & Bioanalysis Services-Preclinical Researchmedicilonz
 
a-rat-pharmacokinetic-pharmacodynamic-model-for-assessment-of-lipopolysacchar...
a-rat-pharmacokinetic-pharmacodynamic-model-for-assessment-of-lipopolysacchar...a-rat-pharmacokinetic-pharmacodynamic-model-for-assessment-of-lipopolysacchar...
a-rat-pharmacokinetic-pharmacodynamic-model-for-assessment-of-lipopolysacchar...Shannon Chesley
 
Alkaline hydrolysis-of-prion-positive-materials-r-murphy
Alkaline hydrolysis-of-prion-positive-materials-r-murphyAlkaline hydrolysis-of-prion-positive-materials-r-murphy
Alkaline hydrolysis-of-prion-positive-materials-r-murphyIIAD
 
Alkaline hydrolysis-of-prion-positive-materials-r-murphy
Alkaline hydrolysis-of-prion-positive-materials-r-murphyAlkaline hydrolysis-of-prion-positive-materials-r-murphy
Alkaline hydrolysis-of-prion-positive-materials-r-murphyIIAD
 
The Importance Of Animal Uses In Animals
The Importance Of Animal Uses In AnimalsThe Importance Of Animal Uses In Animals
The Importance Of Animal Uses In AnimalsJessica Lopez
 
Pre clinical studies
Pre clinical studiesPre clinical studies
Pre clinical studiesKirsha K S
 
Evaluation of the Impact of Biofield Treatment on Physical and Thermal Proper...
Evaluation of the Impact of Biofield Treatment on Physical and Thermal Proper...Evaluation of the Impact of Biofield Treatment on Physical and Thermal Proper...
Evaluation of the Impact of Biofield Treatment on Physical and Thermal Proper...wilhelm mendel
 
Evaluation of the Impact of Biofield Treatment on Physical and Thermal Proper...
Evaluation of the Impact of Biofield Treatment on Physical and Thermal Proper...Evaluation of the Impact of Biofield Treatment on Physical and Thermal Proper...
Evaluation of the Impact of Biofield Treatment on Physical and Thermal Proper...rachelsalk
 
10.1021@ac3033245
10.1021@ac303324510.1021@ac3033245
10.1021@ac3033245Audry Arias
 
Hepatoprotective Activity of Methanolic Extract of Whole Plant of Pulicaria W...
Hepatoprotective Activity of Methanolic Extract of Whole Plant of Pulicaria W...Hepatoprotective Activity of Methanolic Extract of Whole Plant of Pulicaria W...
Hepatoprotective Activity of Methanolic Extract of Whole Plant of Pulicaria W...IOSRJPBS
 
CrossGen-Merck manuscript
CrossGen-Merck manuscriptCrossGen-Merck manuscript
CrossGen-Merck manuscriptKush Sharma
 
International Journal of Engineering and Science Invention (IJESI)
International Journal of Engineering and Science Invention (IJESI)International Journal of Engineering and Science Invention (IJESI)
International Journal of Engineering and Science Invention (IJESI)inventionjournals
 
Renal Toxicity Studies of Albino Rats Exposed to Common Emulsion Paint Solution
Renal Toxicity Studies of Albino Rats Exposed to Common Emulsion Paint SolutionRenal Toxicity Studies of Albino Rats Exposed to Common Emulsion Paint Solution
Renal Toxicity Studies of Albino Rats Exposed to Common Emulsion Paint SolutionPremier Publishers
 
Protective influence of costus afer aqueous extract in rats fed with crude oi...
Protective influence of costus afer aqueous extract in rats fed with crude oi...Protective influence of costus afer aqueous extract in rats fed with crude oi...
Protective influence of costus afer aqueous extract in rats fed with crude oi...Ichipi-ifukor Patrick Chukuyenum
 

Similar to Pig-a (20)

Protective Effect of Taxifolin on Cisplatin-Induced Nephrotoxicity in Rats
Protective Effect of Taxifolin on Cisplatin-Induced Nephrotoxicity in RatsProtective Effect of Taxifolin on Cisplatin-Induced Nephrotoxicity in Rats
Protective Effect of Taxifolin on Cisplatin-Induced Nephrotoxicity in Rats
 
ca3711en.pdf
ca3711en.pdfca3711en.pdf
ca3711en.pdf
 
Jianying Xiao CV 2016
Jianying Xiao CV 2016  Jianying Xiao CV 2016
Jianying Xiao CV 2016
 
IOSR Journal of Pharmacy (IOSRPHR), www.iosrphr.org, call for paper, research...
IOSR Journal of Pharmacy (IOSRPHR), www.iosrphr.org, call for paper, research...IOSR Journal of Pharmacy (IOSRPHR), www.iosrphr.org, call for paper, research...
IOSR Journal of Pharmacy (IOSRPHR), www.iosrphr.org, call for paper, research...
 
Medicilon DMPK & Bioanalysis Services-Preclinical Research
Medicilon DMPK & Bioanalysis Services-Preclinical ResearchMedicilon DMPK & Bioanalysis Services-Preclinical Research
Medicilon DMPK & Bioanalysis Services-Preclinical Research
 
ACUTE, SUB ACUTE & CHRONIC TOXICOLOGICAL STUDIES
ACUTE, SUB ACUTE & CHRONIC TOXICOLOGICAL STUDIESACUTE, SUB ACUTE & CHRONIC TOXICOLOGICAL STUDIES
ACUTE, SUB ACUTE & CHRONIC TOXICOLOGICAL STUDIES
 
a-rat-pharmacokinetic-pharmacodynamic-model-for-assessment-of-lipopolysacchar...
a-rat-pharmacokinetic-pharmacodynamic-model-for-assessment-of-lipopolysacchar...a-rat-pharmacokinetic-pharmacodynamic-model-for-assessment-of-lipopolysacchar...
a-rat-pharmacokinetic-pharmacodynamic-model-for-assessment-of-lipopolysacchar...
 
Alkaline hydrolysis-of-prion-positive-materials-r-murphy
Alkaline hydrolysis-of-prion-positive-materials-r-murphyAlkaline hydrolysis-of-prion-positive-materials-r-murphy
Alkaline hydrolysis-of-prion-positive-materials-r-murphy
 
Alkaline hydrolysis-of-prion-positive-materials-r-murphy
Alkaline hydrolysis-of-prion-positive-materials-r-murphyAlkaline hydrolysis-of-prion-positive-materials-r-murphy
Alkaline hydrolysis-of-prion-positive-materials-r-murphy
 
The Importance Of Animal Uses In Animals
The Importance Of Animal Uses In AnimalsThe Importance Of Animal Uses In Animals
The Importance Of Animal Uses In Animals
 
Pre clinical studies
Pre clinical studiesPre clinical studies
Pre clinical studies
 
Evaluation of the Impact of Biofield Treatment on Physical and Thermal Proper...
Evaluation of the Impact of Biofield Treatment on Physical and Thermal Proper...Evaluation of the Impact of Biofield Treatment on Physical and Thermal Proper...
Evaluation of the Impact of Biofield Treatment on Physical and Thermal Proper...
 
Evaluation of the Impact of Biofield Treatment on Physical and Thermal Proper...
Evaluation of the Impact of Biofield Treatment on Physical and Thermal Proper...Evaluation of the Impact of Biofield Treatment on Physical and Thermal Proper...
Evaluation of the Impact of Biofield Treatment on Physical and Thermal Proper...
 
10.1021@ac3033245
10.1021@ac303324510.1021@ac3033245
10.1021@ac3033245
 
Hepatoprotective Activity of Methanolic Extract of Whole Plant of Pulicaria W...
Hepatoprotective Activity of Methanolic Extract of Whole Plant of Pulicaria W...Hepatoprotective Activity of Methanolic Extract of Whole Plant of Pulicaria W...
Hepatoprotective Activity of Methanolic Extract of Whole Plant of Pulicaria W...
 
CrossGen-Merck manuscript
CrossGen-Merck manuscriptCrossGen-Merck manuscript
CrossGen-Merck manuscript
 
2014 abstracs.pdf
2014 abstracs.pdf2014 abstracs.pdf
2014 abstracs.pdf
 
International Journal of Engineering and Science Invention (IJESI)
International Journal of Engineering and Science Invention (IJESI)International Journal of Engineering and Science Invention (IJESI)
International Journal of Engineering and Science Invention (IJESI)
 
Renal Toxicity Studies of Albino Rats Exposed to Common Emulsion Paint Solution
Renal Toxicity Studies of Albino Rats Exposed to Common Emulsion Paint SolutionRenal Toxicity Studies of Albino Rats Exposed to Common Emulsion Paint Solution
Renal Toxicity Studies of Albino Rats Exposed to Common Emulsion Paint Solution
 
Protective influence of costus afer aqueous extract in rats fed with crude oi...
Protective influence of costus afer aqueous extract in rats fed with crude oi...Protective influence of costus afer aqueous extract in rats fed with crude oi...
Protective influence of costus afer aqueous extract in rats fed with crude oi...
 

Pig-a

  • 1. © The Author 2015. 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. 335 Mutagenesis, 2015, 335–342 doi:10.1093/mutage/gev013 Original Article Advance Access publication 1 May 2015 Original Manuscript Integration of Pig-a, micronucleus, chromosome aberration and comet assay endpoints in a 28- day rodent toxicity study with urethane Leon F. Stankowski, Jr1, *, Marilyn J. Aardema1,2 ,Timothy E. Lawlor1 , Kamala Pant1 , Shambhu Roy1 ,Yong Xu1 and Reem Elbekai1 1 BioReliance Corporation, Rockville, MD 20850, USA, 2 Marilyn Aardema Consulting LLC, Fairfield, OH 45014, USA *To whom correspondence should be addressed. Tel: +1 240 447 2674; Fax: +1 301 738 2362; Email: leon.stankowski@bioreliance.com Received October 1 2014; Revised January 21 2015; Accepted January 21 2015. Abstract As part of the international Pig-a validation trials, we examined the induction of Pig-a mutant reticulocytes and red blood cells (RETCD59− and RBCCD59− , respectively) in peripheral blood of male Sprague Dawley® rats treated with urethane (25, 100 and 250 mg/kg/day) or saline by oral gavage for 29 days. Additional endpoints integrated into this study were: micronucleated reticulocytes (MN-RET) in peripheral blood; chromosome aberrations (CAb) and DNA damage (%tail intensity via the comet assay) in peripheral blood lymphocytes (PBL); micronucleated polychromatic erythrocytes (MN-PCE) in bone marrow; and DNA damage (comet) in various organs at termination (the 29th dose was added for the comet endpoint at sacrifice). Ethyl methanesulfonate (EMS; 200 mg/kg/day on Days 3, 4, 13, 14, 15, 27, 28 and 29) was evaluated as the concurrent positive control (PC). All animals survived to termination and none exhibited overt toxicity, but there were significant differences in body weight and body weight gain in the 250-mg/kg/day urethane group, as compared with the saline control animals. Statistically significant, dose-dependent increases were observed for urethane for: RETCD59− and RBCCD59− (on Days 15 and 29); MN-RET (on Days 4, 15 and 29); and MN-PCE (on Day 29).The comet assay yielded positive results in PBL (Day 15) and liver (Day 29), but negative results for PBL (Days 4 and 29) and brain, kidney and lung (Day 29). No significant increases in PBL CAb were observed at any sample time. Except for PBL CAb (likely due to excessive cytotoxicity), EMS-induced significant increases in all endpoints/tissues.These results compare favorably with earlier in vivo observations and demonstrate the utility and sensitivity of the Pig-a in vivo gene mutation assay, and its ability to be easily integrated, along with other standard genotoxicity endpoints, into 28-day rodent toxicity studies. Introduction The induction and quantification of Pig-a mutant erythrocytes in rodents were first described in 2008 (1–4). Since then, this assay has been the subject of intensive study and global validation coordi- nated by Litron Laboratories and the Genetic Toxicology Technical Committee of the ILSI Health and Environmental Sciences Institute (5–16). These studies indicated that the Pig-a endpoint was highly transferrable and reproducible between laboratories, it could be eas- ily integrated into standard toxicology studies, and that it held great promise as an in vivo mutation assay (17,18). Integration of genotoxicity endpoints into 28-day toxicity studies is included in new regulatory guidelines and protocols, such as those from the International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use [ICH; (19)] and the Organization for Economic Co-operation and Development [OECD; (20–22)], as a way to maximize genotoxicity assessments while minimizing the use of animals. As part of these continuing Pig-a validation trials, we examined the induction of RETCD59− and RBCCD59− in peripheral blood of male Sprague Dawley® rats treated with urethane for 29 consecutive days. For comparison, MN-RET in peripheral blood, chromosome aberrations (CAb) and byguestonJanuary18,2017http://mutage.oxfordjournals.org/Downloadedfrom
  • 2. DNA damage (via the comet assay) in peripheral blood lymphocytes (PBL), micronucleated polychromatic erythrocytes (MN-PCE) in bone marrow and DNA damage in various organs were also evalu- ated. In this study, we demonstrate how every standard in vivo geno- toxicity endpoint, including Pig-a, can be integrated into standard 28-day toxicity studies. Urethane is carcinogenic in multiple rodent species by various routes (23–25), and we use it at BioReliance as a very effective PC in ras H2 transgenic carcinogenicity studies (26). It is also mutagenic and clastogenic in vivo, but generally is considered non-genotoxic in vitro due to its unique metabolism requirements (24,25,27,29). Metabolism of urethane requires CYP2E1 and carboxylesterase isozyme hydrolase A  and proceeds through vinyl carbamate and conversion to an epoxide that can interact with DNA (29,30). Support for this metabolic pathway comes from the observation that urethane-induced MN-PCE frequencies are reduced in CYP2E1-null mice (31). Thus, urethane represents a key test chemical to evaluate the sensitivity and applicability of the Pig-a assay, particularly for the detection of promutagens (18). Materials and Methods Reagents Urethane (CAS no. 51-79-6) and ethyl methanesulfonate (EMS; CAS no. 62-50-0) were purchased from Sigma-Aldrich (St. Louis, MO). Saline for injection (USP; saline) was purchased from B. Braun Medical (Irvine, CA). Prototype MutaFlow® Rat Pig-a Mutation Assay and MicroFlow® Rat Blood Micronucleus Kits were supplied by Litron Laboratories (Rochester, NY). Other reagents and supplies included Lympholyte® - Mammal cell separation media from Cedarlane (Burlington, NC); Anti-PE MicroBeads, LS Columns and a QuadroMACS™ Separator from Miltenyi Biotec (Bergisch Gladbach, Germany); CountBright™ Absolute Count Beads from Invitrogen (Carlsbad, CA). Animals, treatment and harvests The number of animals, procedures and design used for this study were reviewed and approved by the BioReliance Institutional Animal Care and Use Committee and followed the specifications recom- mended in the most current version of The Guide for the Care and Use of Laboratory Animals (32). In addition, the study was conducted in compliance with Good Laboratory Practice regulations (33). Five- to six-weeks old Sprague Dawley® male rats (Harlan; Frederick, MD) were singly housed in polycarbonate cages and acclimated for 11 days prior to the start of dosing. Certified Rodent Diet (Global Diet #2018C; Harlan TEKLAD, Madison, WI) and water were provided ad libitum. Sani-Chip Hardwood bedding (P.J. Murphy Forest Products, Montville, NJ) was used to absorb liquids. Animals were housed in an environmentally controlled room with temperatures between 21 and 24°C,a relative humidity of 30–70%,at least 10 changes of fresh HEPA- filtered air every hour, and a 12-h light/12-h dark cycle. Animals were 7–8-weeks old at the start of dosing and weighed 176–225 g. General daily clinical observations, as well as weekly detailed observations, were performed for each animal. Body weight and food consumption were monitored throughout the treatment period. Animals were treated for 29 consecutive days with urethane at doses of 25, 100 and 250 mg/kg/day, or the saline vehicle control (all n = 10; Table 1). The EMS PC was administered at a dose of 200 mg/ kg/day on Days 3, 4, 13, 14, 15, 27, 28 and 29 (n = 6). The final dose administered on Day 29 was necessary for the comet endpoint. Urethane dose formulations were prepared twice (on Days 0 and 14), aliquoted for daily use, stored at 2–8ºC and maintained at room temperature for at least 30 min prior to use. EMS dose formulations were freshly prepared and maintained at room temperature until use. All dose formulations were prepared in saline, corrected for potency and administered by oral gavage using a 10-ml/kg dose volume, with the first day of dosing designated as Day 1. Blood and tissue collection Blood for the various endpoints was collected from the retro-orbital sinus under 70% CO2 /30% O2 anesthesia at the volumes and time points indicated (Table 2). Each collection was ~2.5–3 h after the appro- priate daily dose as is typical for the comet endpoint (22). Blood for the comet and the CAb assays was collected directly into sodium heparin tubes, whereas blood for the Pig-a and MN-RET assays was initially drawn into an empty tube and then quickly aliquoted into the appropri- ate volume of heparin (Litron kit supplied). Samples also were collected for clinical pathology (Clin Path) analysis (hematology, clinical chemis- try), as is standard for a 28-day rodent toxicity assay (not shown). All animals were sacrificed by CO2 overdose on Day 29, immedi- ately after the final blood collections, and a complete necropsy was performed. The left kidney, and consistent portions of the left lateral liver lobe, brain, spleen and lungs were collected for the comet assay. Similarly, the left femur was reserved for bone marrow micronucleus assessment. All remaining portions of these tissues, as well as numer- ous other tissues and the entire carcass including animal identifica- tion, were collected and fixed in 10% neutral buffered formalin for histopathologic evaluation (as in a standard 28-day rodent toxicity assay; data not shown). Pig-a assay Blood was processed, labeled and analysed using Prototype In Vivo MutaFlow kits and instructions (version 120209, with minor Table 1.  Study design Group Treatment Animals per groupa Dose level Dose formulation (mg/kg/day) (mg/ml) 1 Saline 10 0.00 0.00 2 Urethane 10 25.0 2.50 3 Urethane 10 100 10.0 4 Urethane 10 250 25.0 5 EMS 6 200 20.0 a Males only. Ten animals per group were treated with urethane or the ve- hicle control (as per a standard 28-day rodent toxicity study). Eight animals/ group (or all from the EMS positive control group) initially were analysed prior to dosing to determine the pre-dose Pig-a mutant frequencies. One ani- mal from the 100 mg/kg/day urethane group was excluded from all subsequent genetic toxicology endpoint analyses (due to a high spontaneous Pig-a mutant frequency), and the first six animals remaining in each group were analysed for all genetic toxicology endpoints. Table 2.  Sampling times and blood volumes (µl)a Day N Pig-a MN-RET CAb Comet Clin path Total −5 or −3 8 100 – – – – 100 4 6 – 150 250 100 – 500 15 6 100 150 250 100 – 600 29 6 100 150 250 100 800 1400 a Blood was collected ~2.5–3 h after dosing, except for the pre-dose samples on Days −3 or −5. 336 L. F. Stankowski et al., 2015, Vol. 30, No. 3 byguestonJanuary18,2017http://mutage.oxfordjournals.org/Downloadedfrom
  • 3. modifications) and with immunomagnetic enrichment (34). A stand- ard template was used and data was acquired using a BD FACSCanto II flow cytometer with FACSDiva Software (BD Biosciences; San Jose, CA). Whenever possible, ≥100 million total RBCs and ≥3 mil- lion RETs were analysed. For each animal, the total number of RET and RBC equivalents analysed, as well as the number and frequency of RETCD59− and RBCCD59− , were determined/calculated. MN-RET assay Blood was processed, labeled and analysed according to the instruc- tion manual (version 120702)  received with the MicroFlow® Rat Blood Micronucleus Kit. Approximately 20 000 RET were scored for micronuclei, per animal and time point, where possible. The same flow cytometer and software were used for data acquisition and analysis. MN-PCE assay Bone marrow was aspirated by syringe, transferred to a centrifuge tube containing cold fetal bovine serum (FBS), pelleted by centrifu- gation, resuspended in a small volume of FBS and spread onto a glass slide (2/animal). The slides were air dried for 24–72 h, fixed in methanol and stained with acridine orange for microscopic evalua- tion. Coded slides were scored by fluorescent microscopy at 1000× magnification for the presence of micronuclei (2000 PCEs/animal; 12 000 per group), and for the proportion of PCEs to total eryth- rocytes (PCE/EC ratio) as a measure of bone marrow cytotoxicity (1000 total erythrocytes/animal). Ex vivo PBL CAb assay The ex vivo PBL CAb assay was performed and scored as pre- viously reported (35). Briefly, whole blood was washed by cen- trifugation using phosphate-buffered saline containing ~2% (v/v) FBS and resuspended in a 10-fold volume of RPMI 1640 medium containing 25-mM HEPES, 10% (v/v) heat inactivated FBS, 100- units/ml penicillin, 100-μg/ml streptomycin, 2-mM L-glutamine and 20-μg/ml phytohemagglutinin-P to stimulate lymphocyte proliferation. Single cultures were established in 25-cm2 flasks (one per animal) and incubated at 37 ± 2°C in a humidified atmos- phere of ~5% CO2 in air. Cultures were grown for ~48 h, with Colcemid® (0.2 μg/ml) being added for the last ~2.5 h before har- vest. Duplicate slides were prepared from each culture, stained with Giemsa, air dried and coded, and 100 cells with 2n ± 2 chro- mosomes were scored per animal for structural aberrations and numerical aberrations, whereas 500 cells/ animal were evaluated for mitotic index. Comet assay The alkaline comet assay was used to analyze all tissues using the latest JaCVAM protocol (36,37). Peripheral blood samples were centrifuged and resuspended in phosphate-buffered saline to pre- pare comet slides. Liver, kidney, lung, brain and spleen were placed in cold mincing buffer and finely minced with a pair of fine scis- sors to release the cells. Each cell suspension was strained through a 40-µm sieve into a 50-ml-conical tube and stored on wet ice until the slides were prepared. Approximately 2.5–5 µl of cell suspension were mixed with 75-µl 0.5% (w/v) low melting agarose and applied to glass microscope slides (four slides/animal/tissue) that were previ- ously coated with 1% (w/v) normal melting agarose (to minimize sample degradation, dosing and necropsy was staggered and highly choreographed, such that processing up to the application to the slides was accomplished in ~15 min or less). The slides were kept at 2–8°C for at least 10 to 15 min to solidify. Following solidification, the slides were submerged in cold lysis solution (100-mM Na2 EDTA, 2.5-M NaCl, 10-mM Tris, 1% Triton X-100 and 10% dimethyl sulfoxide; pH 10) for at least overnight at 2–8°C. After lysis, slides were neutralized in 0.4-M Tris (pH ~7.5), transferred to the electrophoresis chamber which was slowly filled with cold electrophoresis buffer (300-mM NaOH and 1-mM Na2 EDTA; pH 13), and maintained for 20 min at 2–10°C, protected from light, to allow the DNA to unwind. Electrophoresis was con- ducted for 30 min at 0.7 V/cm, at 2–10°C and protected from light. After electrophoresis, the slides were removed and washed with neu- tralization buffer, dehydrated with 200-proof ethanol, air dried and stored at room temperature with desiccant. Slides were stained with Sybr-gold™ in Tris-boric acid- EDTA buffer. Up to 50 randomly selected, nonoverlapping cells were scored per slide, for a total of 100 cells evaluated per animal and tissue, using the comet Assay IV scoring system (version 4.11; Perceptive Instruments Ltd.; Bury St. Edmunds, UK). DNA damage was assessed based upon %Tail DNA. Each slide was also examined for indications of possible cytotoxicity (i.e. the percentage of “clouds” in 100 cells/slide, where possible). Data analyses Pig-a analyses were performed on log(10) transformed mutant frequencies (RETCD59− and RBCCD59− ), and untransformed %RETs, using the individual animal as the unit of measure. An offset of 0.1 was also added to each mutant RET value prior to transformation to avoid zero values. One-way analysis of variance was performed (for mutant frequencies and %RETs), followed by Dunnett’s post hoc analysis to compare each dose to the vehicle control. A pair- wise t-test was performed to compare the saline and EMS PCs. Significance for all statistical analyses was established at the P  0.05 or 0.01 levels using a two-tailed test. The percent MN-RET and %RET per group were tested for nor- mal distribution (Levene’s test, P  0.05), and log transformed if not normally distributed. Urethane data were analysed using one-way analysis of variance followed by Dunnett’s post hoc analysis. A pair- wise t-test was performed to compare saline versus the EMS PC. MN-PCE frequencies were tested for normal distribution (Levene’s test, P  0.05; and log transformed if not normally distrib- uted) and analysed using the binomial distribution (Kastenbaum– Bowman tables). CAb data were analysed using Fisher’s exact test, while the comet assay results (animal mean calculated from the median %Tail DNA, 50 cells/slide per animal) were analysed as described for MN-RET. Body weight, food consumption and organ weight data were analysed by Dunnett’s test. Results Survival and clinical observations All animals survived to Day 29 and none exhibited overt signs of toxicity due to urethane or EMS treatment (data not shown). The dose level for the EMS PC is the same as we normally use in an acute, stand-alone comet assay, or in a combination comet/micro- nucleus study using three daily dose administrations. However, the cumulative dose and bone marrow toxicity induced by the eight administrations used here was clearly cytotoxic to the bone mar- row, as evidenced by the %RET, PCE/EC, and CAb results below. In contrast, there were no significant changes in Clin Path for the Integration of Pig-a, micronucleus, chromosome aberration and comet assay endpoints, 2015, Vol. 30, No. 3 337 byguestonJanuary18,2017http://mutage.oxfordjournals.org/Downloadedfrom
  • 4. urethane-treated animals except for an increase in % monocytes at a dose of 250 mg/kg/day. Body weight, body weight change and food consumption Significant differences in body weight were noted for the 250-mg/kg/ day urethane treatment group, as well as the EMS PC group, starting on Day 8 (P  0.05; not shown). In addition, significant differences in body weight gain were noted during the first 2 weeks of the study, for the same two groups. Absolute mean body weight gains over the course of the study, as compared to the control animals, were 22.9% and 51.4% lower, respectively (not shown). Group mean food con- sumption values were lower in the 250-mg/kg/day urethane group, as compared to the control animals, but the differences were not significant (P 0.05; food consumption data were not collected for the EMS PC group). Pig-a assay Statistically significant, dose-dependent increases in the frequency of RETCD59− were observed at doses of 100 and 250 mg/kg/day on Days 15 and 29 (P  0.05; Figure 1, Table 3). The maximum observed RETCD59− frequencies were ~74- and 24-fold vehicle control values on Days 15 and 29, respectively (the relatively smaller increase on Day 29 was due to the higher mutant frequency in the vehicle control animals at the later time point). Significant increases in RBC CD59− also were observed at a dose of 250 mg/kg/day on Day 15, and at doses of 100 and 250 mg/kg/day on Day 29 (P  0.05; Figure 2, Table 3). These increases reached maximums of ~4.2- and 8.1-fold control values on Days 15 and 29, respectively. MN-RET assay %RET, measured concurrently with the MN-RET endpoint, indi- cated urethane and the EMS PC reached the bone marrow and induced significant toxicity. Statistically significant decreases in %RET were observed for urethane at a dose of 250 mg/kg/day on Days 4 and 29 (but not Day 15), and for EMS on Days 15 and 29 (Figure 3, Table 3). The apparent lack of bone marrow toxicity for urethane on Day 15 is curious and may reflect an initial compensa- tory increase in erythropoiesis. Statistically significant, dose-dependent increases in the frequency of MN-RET, to ~4.0- to 6.6-fold control values, were observed for urethane at all doses on Days 4 and 29, and doses of 100 and 250 mg/kg/day on Day 15 (P  0.05 or 0.01; Figure 4, Table 3). EMS also induced significant increases in MN-RET at all time points; these increases were larger at the later two time points, likely due to the change in dose regimen (two vs. three daily doses before the scheduled collection times). MN-PCE assay Significant, dose-dependent increases in MN-PCE were observed for urethane-treated animals at terminal sacrifice at a dose of 250 mg/ kg/day (Figure 5; Table 3). EMS induced a much larger increase in MN-PCE. In contrast to measurements in the peripheral blood, sig- nificant decreases in PCE/EC ratios were only noted for EMS (not shown). Figure 1.  Frequency of RETCD59− induced by urethane or the EMS PC (all n = 6; ±1 SD). Statistically significant increases are indicated at P  0.05 and 0.01 levels (* and **, respectively). The 95% upper control limit for historical negative control RETCD59− data was 2.90 × 10−6 . Table 3.  Summary of results Endpoint Maximum fold-increasea Day 4 Day 15 Day 29 RETCD59− nd 74 24 RBCCD59− nd 4.2 8.1 MN-RET 6.6 4.0 5.3 MN-PCE nd nd 5.0 PBL cab – – – Comet PBL – 3.0 – Brain nd nd – Kidney nd nd – Liver nd nd 1.5 Lung nd nd – Spleen nd nd – nd = not determined. – = negative response. a Only the greatest observed increase, at any dose level or time point, is reported; any numeric entry represents a statistically significant, positive re- sponse (P  0.05). Figure  2.  Frequency of RBCCD59− induced by urethane or the EMS PC (all n = 6; ±1 SD). Statistically significant increases are indicated at P  0.05 and 0.01 levels (* and **, respectively).The 95% upper control limit for historical negative control RBCCD59− data was 1.39 × 10−6 . 338 L. F. Stankowski et al., 2015, Vol. 30, No. 3 byguestonJanuary18,2017http://mutage.oxfordjournals.org/Downloadedfrom
  • 5. Ex vivo PBL CAb assay No significant increases were observed in structural or numerical aberrations at any dose or sample time for the urethane-treated ani- mals (results not shown, but summarized in Table 3). This lack of response was clearly not due to a lack of exposure, since relative mitotic indices decreased in a dose dependent manner and reached relative mitotic indices of ~69, 47 and 53% at a dose of 250 mg/kg/ day on Days 4, 15 and 29, respectively. Cultures from EMS-treated animals were unable to be scored due to insufficient metaphases, likely as the result of excessive toxicity (relative mitotic indices were ~17, 10 and 9% of control values on Days 4, 15 and 29, respectively). Comet assay Statistically significant, dose-dependent increases in median %Tail DNA were observed for urethane only in PBL at Day 15, and in liver at terminal sacrifice (P  0.05; Figures 6 and 7; Table 3). Urethane was considered to be negative in all other tissues and time points analysed.The EMS PC induced significant increases in median %Tail DNA in all tissues and at all sampling times (P  0.05). The percent- age of clouds observed in the urethane-treated animals was compa- rable to, or less than, that observed in the vehicle control animals, for all tissues and time points (not shown). In contrast, increases in the percentage of clouds in the EMS-treated animals ranged from ~2- to 4-fold (blood on Day 4 and brain, lung and spleen) to ~8-fold or greater in the other tissues, and blood at the later time points (not shown). Discussion One goal of this study was to validate the integration of the in vivo Pig-a assay, as well as other genetic toxicology endpoints, into a typical 28-day rodent toxicity assay in compliance with Good Laboratory Practice regulations (33). As such, all of the typical body and organ weight, cage-side and detailed observations, food con- sumption and Clin Path procedures/analyses were performed (tissues were collected for histopathology but not analysed due to the time and expense involved). Only the results relevant to interpretation of the effects of urethane are reported here. Based on data in the literature, urethane was positive in most in vivo cytogenetic and mutation studies in vivo (24,25,28). Results from our study compare favorably with these previous in vivo obser- vations as discussed below. Urethane induced statistically significant, dose-dependent increases in the frequencies of MN-RET at all time points analysed, and in MN-PCE at terminal sacrifice. The maximal fold-increases observed were similar (~4- to 6-fold for all observa- tions), even though the absolute frequencies were quite different for MN-RET and MN-PCE. It should be noted that positive (significant) responses were observed for MN-RET at doses ≥100 mg/kg/day, but only at 250 mg/kg/day for the MN-PCE endpoint. This difference is likely due to the improved sensitivity and power afforded by the ability to routinely score many more cells using the automated flow cytometric method (38). Also, the apparent “disappearance” of the positive MN-RET response at a dose of 25.0 mg/kg/day on Day 15 may reflect that the dose/response was at or near the limit of detec- tion for the assay. Urethane also induced statistically significant, dose-dependent increases in the comet assay (median %Tail DNA) in PBL on Day 15, and in liver at terminal sacrifice. However, the relative increases (1.5- to 3.0-fold control values) were even less than observed for MN-RET and MN-PCE. In addition, no significant increases were observed for the comet endpoint in blood on Day 4 or 29. Thus, the Figure 3.  Percent RET in peripheral blood, following treatment with urethane or the EMS PC, as assessed by flow cytometry (all n = 6; ±1 SD). Figure  4.  Frequency of MN-RET induced by urethane or the EMS PC (all n = 6; ±1 SD). Statistically significant increases are indicated at P  0.05 and 0.01 levels (* and **, respectively).The 95% upper control limit for historical negative control MN-RET data was 0.175%. Figure  5.  Frequency of MN-PCE induced by urethane or the EMS PC (all n = 6; ±1 SD). Statistically significant increases are indicated at P  0.05 and 0.01 levels (* and **, respectively).The 95% upper control limit for historical negative control MN-PCE data was 0.0947%. Integration of Pig-a, micronucleus, chromosome aberration and comet assay endpoints, 2015, Vol. 30, No. 3 339 byguestonJanuary18,2017http://mutage.oxfordjournals.org/Downloadedfrom
  • 6. “positive” comet response, especially in blood, is considered to be suspect. An apparent dose-dependent increase in %Tail DNA was observed in lung, but the individual values did not reach statisti- cal significance (P 0.05). Although dose-dependent decreases in median %Tail DNA were observed in the brain, the assay condi- tions were not optimized for detection of crosslinkers nor was that intended for this study. The lack of any appreciable increases in clouds and the lack of any significant deleterious changes in Clin Path parameters suggest that any significant increases in comet response were not due to cytotoxicity. Also to our knowledge, this study represents the first reported in vivo comet assay with urethane. The negative comet results in lung are perhaps unexpected, since it is one of the principal sites for tumor formation (24,25), and positive results have been observed in transgenic rodent mutation (TRM) assays, in multiple tissues includ- ing forestomach, liver and lung, following oral dosing [reviewed in refs. (39–41)]. However, in the cases of earlier positive TRM results, the cumulative dose levels generally were much higher, and/or the sampling time was much later, than studied here (see additional discussion below regarding cumulative dose and sampling times). Likewise, the sampling time was our standard, in compliance with OECD recommendations (22), but not necessarily optimal based upon uptake and metabolism kinetics or DNA repair. No significant increases were observed for urethane in the CAb assay at any dose or sampling time. In searching the literature, we found no reports of structural CAb induced by urethane when scored by metaphase analysis; rather, the induction of CAb was inferred using micronuclei as a surrogate for chromosome breakage. Although the micronucleus test should be considered the in vivo test of choice for CAb (21), little additional effort was required to add the CAb endpoint to this study, as has been suggested elsewhere (42). Since a daily dosing regimen was used, and the 48-h culture time after stimulation has proved reliable in this laboratory, the recom- mended sampling time for the comet assay that was used here (22) is likely not responsible for the lack of a positive CAb response for urethane. However, we also note the PCs for this assay failed, but likely due to excessive toxicity. Finally, and most importantly for this study, urethane induced statistically significant, dose-dependent increases in the frequen- cies of RETCD59− and RBCCD59− at both time points analysed. Larger responses were observed in RETCD59− , which is consistent with earlier observations for other agents and is due to slower turnover in the RBC population (18). The maximum mutant frequencies observed at a dose of 250 mg/kg/day were ~20 × 10−6 for RETCD59− on Day 15, and ~6 × 10−6 for RBCCD59− on Day 29. These numbers compare very favorably to those observed for urethane used under identical con- ditions as a PC in a study of methyl carbamate (43). The relatively larger fold-increase in RETCD59− on Day 15 versus Day 29 (Table 3) is the result of an approximate 3-fold difference in negative con- trol mutant frequencies at those time points (2.82 vs. 0.678 × 10−6 , respectively). The 24- and 74-fold increases in RETCD59− frequencies on Day 29 and 15 far exceeded those observed in the other positive endpoints. Based upon the biology and expression time for the RETCD59− and RBCCD59− to appear in the peripheral blood, and as seen for a num- ber of chemicals, at least some additional increases in mutant fre- quencies could be expected (5,6,18,44,45). Given sufficient time, it is anticipated that the fold-increases in RBCCD59− frequencies would reach levels similar to those observed for RETCD59− . These results, in comparison to the other endpoints studied, further reinforce the dramatic benefits of repeat dosing, and accumulation of induced mutants, that are associated with the Pig-a assay [as has been reported previously and reviewed by Gollapudi et al. (18)]. Urethane has been reported to be one of the weakest mutagens examined to date in the TRM assays using the currently recom- mended study design (46–48). Only half of the urethane treatments previously reviewed (28/56) produced positive findings [by any route; results compiled in refs. (39–41)]. Although mutant frequen- cies in the lung and liver reached ~5- to 19-fold control values in some of these TRM studies (representing induced mutant frequen- cies of ~800–1300 × 10‒6 ), these were 6- to 8-month drinking water or feed studies performed over much longer intervals (105–237 days) at ~2- to 10-fold higher cumulative doses (13 650–69 678 mg/kg) than in our study (7250 mg/kg). Most other oral dosing TRM studies (gavage or diet) reported in the literature using dose regimens similar to our study (1400–5600 mg/kg by oral gavage) resulted in approxi- mately only 2-fold increases in mutant frequencies in the lung or liver (representing induced mutant frequencies of ~60–110 × 10−6 ) or negative results (39–41). Similar TRM studies (cumulative doses Figure  7.  Comet analysis in various tissues following 29 daily doses of urethane (median %Tail DNA ±1 SD; n  =  6, except as noted). EMS PC responses were off scale (~30–34%). Statistically significant increase indicated at P  0.05 level (*). Lung results from one animal were excluded as an extreme outlier (13.1%). The 95% upper control limits for historical negative control median %Tail DNA data were: 2.48% (liver), 22.3% (brain), 4.20% (spleen), 4.14% (kidney) and 11.6% (lung). Figure 6.  Comet analysis in PBL following 29 daily doses of urethane (median %Tail DNA; all n = 6; ±1 SD). EMS PC responses were off scale (~16–26%). Statistically significant increases are indicated at P  0.01 level (**).The 95% upper control limit for historical negative control median %Tail DNA data was 0.750%. 340 L. F. Stankowski et al., 2015, Vol. 30, No. 3 byguestonJanuary18,2017http://mutage.oxfordjournals.org/Downloadedfrom
  • 7. of 700 to 5600 mg/kg by oral gavage) produced ~1.7- to 2.8-fold increases in mutant frequencies in haematopoietic tissues (represent- ing induced mutant frequencies of ~35–100 × 10−6 ). Although the net increases in Pig-a mutant frequencies reported here were smaller than some observed in the TRM assays, the fold-increases were larger due to the lower spontaneous mutant frequencies for Pig-a. Also as previously stated, the Pig-a mutant frequencies would be expected to increase with a longer phenotypic expression time. These results demonstrate the utility and sensitivity of the Pig-a in vivo gene mutation assay. This endpoint, as well as the other genetic toxicology endpoints, can be integrated into repeat dose tox- icity studies easily and economically, resulting in a comprehensive analysis of genotoxicity with reduced animal use. The relative sensi- tivity of the various endpoints is likely due in part to the statistical power and sample sizes of the various assays, and the ability of Pig-a mutant cells to persist and accumulate during repeated dosing. The negative and/or weak CAb and comet responses are simi- lar to our previous observations for 4-nitroquinoline oxide using a 29-day dose regimen (16). Those results, and the weak MN-PCE and comet responses here, further demonstrate that not all genotoxicity endpoints respond similarly. Although it is certainly possible to inte- grate multiple (or all of these) endpoints into a single study, it is not necessarily prudent or scientifically justified. As previously discussed (45), the Pig-a and MN-RET are easily integrated, complimentary endpoints that offer the ability to detect gene mutation and cytoge- netic damage, sometimes arising from two distinct modes of action, that cannot be universally detected for all agents by a single assay. Where limitations exist (e.g. questions over target tissue exposure), then the comet assay would be a valuable addition. Thus, care must be taken in dose, endpoint and tissue selection when incorporating genotoxicity endpoints into routine toxicity studies as has been rec- ommended, for example, by the ICH (19), the OECD (20–22) and the US Food and Drug Administration (42). Funding This work was supported by BioReliance Corporation and also sup- ported in part by a grant to Litron Laboratories (National Institute of Health/National Institute of Environmental Health Sciences, R44ES018017). Acknowledgements The authors thank Sarah Miller for her expert technical assistance, and Stephen Dertinger for invaluable advice and support. The contents herein are solely the responsibility of the authors, and do not necessarily represent the official views of the institutions involved. Conflict of interest statement: All of the authors are employees of BioReliance Corporation and M.J.A. also serves as a consultant to BioReliance. BioReliance is a contract research organization offering these and other studies for sale. References 1. Bryce, S. M., Bemis, J. C. and Dertinger, S. D. (2008) In vivo mutation assay based on the endogenous Pig-a locus. Environ. Mol. Mutagen., 49, 256–264. 2. Miura, D., Dobrovolsky, V. N., Kasahara, Y., Katsuura, Y. and Heflich, R. H. (2008) Development of an in vivo gene mutation assay using the endogenous Pig-A gene: I. Flow cytometric detection of CD59-negative peripheral red blood cells and CD48-negative spleen T-cells from the rat. Environ. Mol. Mutagen., 49, 614–621. 3. Miura, D., Dobrovolsky, V. N., Mittelstaedt, R. A., Kasahara, Y., Katsuura, Y. and Heflich, R. H. (2008) Development of an in vivo gene mutation assay using the endogenous Pig-A gene: II. Selection of Pig-A mutant rat spleen T-cells with proaerolysin and sequencing Pig-A cDNA from the mutants. Environ. Mol. Mutagen., 49, 622–630. 4. Phonethepswath, S., Bryce, S. M., Bemis, J. C. and Dertinger, S. D. (2008) Erythrocyte-based Pig-a gene mutation assay: demonstration of cross-spe- cies potential. Mutat. Res., 657, 122–126. 5. Phonethepswath, S., Franklin, D., Torous, D. K. et al. (2010) Pig-a muta- tion: kinetics in rat erythrocytes following exposure to five prototypical mutagens. Toxicol. Sci., 114, 59–70. 6. Dertinger, S. D., Phonethepswath, S., Franklin, D. et al. (2010) Integration of mutation and chromosomal damage endpoints into 28-day repeat dose toxicology studies. Toxicol. Sci., 115, 401–411. 7. Bhalli, J. A., Shaddock, J. G., Pearce, M. G., Dobrovolsky, V. N., Cao, X., Heflich, R. H. and Vohr, H. W. (2011) Report on stage III Pig-a mutation assays using benzo[a]pyrene. Environ. Mol. Mutagen., 52, 731–737. 8. Bhalli, J. A., Pearce, M. G., Dobrovolsky, V. N. and Heflich, R. H. (2011) Manifestation and persistence of Pig-a mutant red blood cells in C57BL/6 mice following single and split doses of N-ethyl-N-nitrosourea. Environ. Mol. Mutagen., 52, 766–773. 9. Cammerer, Z., Bhalli, J. A., Cao, X. et al. (2011) Report on stage III Pig-a mutation assays using N-ethyl-N-nitrosourea-comparison with other in vivo genotoxicity endpoints. Environ. Mol. Mutagen., 52, 721–730. 10. Dertinger, S. D., Phonethepswath, S., Weller, P. et al. (2011) International Pig-a gene mutation assay trial: evaluation of transferability across 14 laboratories. Environ. Mol. Mutagen., 52, 690–698. 11. Dertinger, S. D., Phonethepswath, S., Weller, P. et al. (2011) Interlaboratory Pig-a gene mutation assay trial: studies of 1,3-propane sultone with immu- nomagnetic enrichment of mutant erythrocytes. Environ. Mol. Mutagen., 52, 748–755. 12. Kimoto, T., Chikura, S., Suzuki, K. et al. (2011) Further development of the rat Pig-a mutation assay: measuring rat Pig-a mutant bone marrow erythroids and a high throughput assay for mutant peripheral blood retic- ulocytes. Environ. Mol. Mutagen., 52, 774–783. 13. Lemieux, C. L., Douglas, G. R., Gingerich, J., Phonethepswath, S., Torous, D. K., Dertinger, S. D., Phillips, D. H., Arlt, V. M. and White, P. A. (2011) Simultaneous measurement of benzo[a]pyrene-induced Pig-a and lacZ mutations, micronuclei and DNA adducts in Muta™ Mouse. Environ. Mol. Mutagen., 52, 756–765. 14. Lynch, A. M., Giddings, A., Custer, L., Gleason, C., Henwood, A., Aylott, M. and Kenny, J. (2011) International Pig-a gene mutation assay trial (stage III): results with N-methyl-N-nitrosourea. Environ. Mol. Mutagen., 52, 699–710. 15. Shi, J., Krsmanovic, L., Bruce, S. et al. (2011) Assessment of genotoxicity induced by 7,12-dimethylbenz(a)anthracene or diethylnitrosamine in the Pig-a, micronucleus and Comet assays integrated into 28-day repeat dose studies. Environ. Mol. Mutagen., 52, 711–720. 16. Stankowski, L. F. Jr, Roberts, D. J., Chen, H., Lawlor, T., McKeon, M., Murli, H., Thakur, A. and Xu, Y. (2011) Integration of Pig-a, micronu- cleus, chromosome aberration, and Comet assay endpoints in a 28-day rodent toxicity study with 4-nitroquinoline-1-oxide. Environ. Mol. Muta- gen., 52, 738–747. 17. Schuler, M., Gollapudi, B. B., Thybaud, V. and Kim, J. H. (2011) Need and potential value of the Pig-ain vivo mutation assay-a HESI perspective. Environ. Mol. Mutagen., 52, 685–689. 18. Gollapudi, B.B., Lynch, A.M., Heflich, R.H. et  al. (2014) The in vivo Pig-a mutant assay: a report of the International Workshop on Geno- toxicity Testing (IWGT) workgroup. Mutat. Res. doi:10.1016/j.mrgen- tox.2014.09.007. 19. International Conference on Harmonization. (2008) Guidance on geno- toxicity testing and data interpretation for pharmaceuticals intended for human use S2(R1) (09 November 2011). Available at: http://www.ich.org/ fileadmin/Public_Web_Site/ICH_Products/Guidelines/Safety/S2_R1/Step4/ S2R1_Step4.pdf (accessed 21 January 2015). 20. Organisation of Economic Co-operation and Development. (2014) Guide- line for Testing of Chemicals: Test Guideline 474 - Mammalian Erythro- cyte Micronucleus Test, adopted 26 September 2014, OECD, Paris. Avail- Integration of Pig-a, micronucleus, chromosome aberration and comet assay endpoints, 2015, Vol. 30, No. 3 341 byguestonJanuary18,2017http://mutage.oxfordjournals.org/Downloadedfrom
  • 8. able at: http://www.oecd-ilibrary.org/docserver/download/9714541e.pdf (accessed 21 January 2015). 21. Organisation of Economic Co-operation and Development. (2014) Guideline for Testing of Chemicals: Test Guideline 475 - Mammalian Bone Marrow Chromosomal Aberration Test, adopted 26 September 2014, OECD, Paris. Available at: http://www.oecd-ilibrary.org/docserver/ download/9714551e.pdf (accessed 21 January 2015). 22. Organisation of Economic Co-operation and Development. (2014) Guide- line for Testing of Chemicals: Test Guideline 489 - In Vivo Mammalian Alkaline Comet Assay, adopted 26 September 2014, OECD, Paris. Avail- able at: http://www.oecd-ilibrary.org/docserver/download/9714511e.pdf (accessed 21 January 2015). 23. Salmon, A.G. and Zeise, L. (1991) Risks of Carcinogenesis from Urethane Exposure. CRC Press, Boca Raton, FL. 24. International Agency for Research on Cancer. (2010). Ethyl carbamate (urethane). IARC Monogr. Carcinog, Risks, Hum, 96, 1281–1307. 25. European Commission. (2012) Recommendation from the Scientific Com- mittee on Occupational Exposure Limits for Ethyl Carbamate [Urethane]. SCOEL/SUM/172. Available at http://ec.europa.eu/social/BlobServlet?doc Id=7723langId=en (accessed 21 January 2015). 26. Shah, S. A., Paranjpe, M. G., Atkins, P. I. and Zahalka, E. A. (2012) Reduc- tion in the number of animals and the evaluation period for the positive control group in Tg.rasH2 short-term carcinogenicity studies. Int. J. Toxi- col., 31, 423–429. 27. Sotomayor, R. E. and Collins, T. F. (1990) Mutagenicity, metabolism, and DNA interactions of urethane. Toxicol. Ind. Health, 6, 71–108. 28. Tweats, D.J., Blakey, D., Heflich, R.H. et al. (2007) Report of the IWGT working group on strategy/interpretation of regulatory in vivo tests II. Identification of in vivo-only positive compounds in the bone marrow micronucleus test. Mutat. Res. 627, 92–105. 29. Forkert, P. G. and Lee, R. P. (1997) Metabolism of ethyl carbamate by pulmonary cytochrome P450 and carboxylesterase isozymes: involvement of CYP2E1 and hydrolase A. Toxicol. Appl. Pharmacol., 146, 245–254. 30. Guengerich, F. P. and Kim, D. H. (1991) Enzymatic oxidation of ethyl car- bamate to vinyl carbamate and its role as an intermediate in the formation of 1,N6-ethenoadenosine. Chem. Res. Toxicol., 4, 413–421. 31. Hoffler, U., Dixon, D., Peddada, S. and Ghanayem, B. I. (2005) Inhibition of urethane-induced genotoxicity and cell proliferation in CYP2E1-null mice. Mutat. Res., 572, 58–72. 32. National Research Council, Institute of Laboratory Animal Resources Commission on Life Sciences. (2011) Guide for the Care and Use of Labo- ratory Animals. National Academy Press, Washington, DC. 33. United States Food and Drug Administration. (1987) Good Laboratory Practice for Nonclinical Laboratory Studies. Code of Federal Regulations, Title 21, Part 58. Federal Register, 52, 33768–33782. 34. Dertinger, S. D., Bryce, S. M., Phonethepswath, S. and Avlasevich, S. L. (2011) When pigs fly: immunomagnetic separation facilitates rapid deter- mination of Pig-a mutant frequency by flow cytometric analysis. Mutat. Res., 721, 163–170. 35. Erexson, G.L., Farabaugh, C.S., Yung, K.M., and Stojhovic, G.P. 2004. An in vivo-in vitro study protocol for the conduct of the rat peripheral blood lymphocyte (RPBL) chromosome aberrations (CA) assay. Environ. Mol. Mutagen., 44, 198. 36. Validation Management Team (VMT) coordinated by JaCVAM (Japanese Committee for the Validation of the Alternative Methods). (2009) Inter- national Validation of the In Vivo Rodent Alkaline Comet Assay for the Detection of Genotoxic Carcinogens (Protocol Version 14.2.). Available at http://www.oecd.org/officialdocuments/publicdisplaydocumentpdf/?cote= ENV/JM/MONO(2014)10doclanguage=en (accessed 21 January 2015). 37. Rothfuss, A., O’Donovan, M., De Boeck, M. et al. (2010) Collaborative study on fifteen compounds in the rat-liver Comet assay integrated into 2- and 4-week repeat-dose studies. Mutat. Res., 702, 40–69. 38. Kissling, G. E., Dertinger, S. D., Hayashi, M. and MacGregor, J. T. (2007) Sensitivity of the erythrocyte micronucleus assay: dependence on number of cells scored and inter-animal variability. Mutat. Res., 634, 235–240. 39. Organisation for Economic Cooperation and Development. (2009) Detailed Review Paper on Transgenic Rodent Mutation Assays, Series on Testing and Assessment, N° 103. ENV/JM/MONO(2009)7. OECD, Paris. Available at: http://www.oecd.org/officialdocuments/publicdisplaydocume ntpdf/?cote=env/jm/mono(2009)7doclanguage=en (accessed 21 January 2015). 40. Organisation for Economic Cooperation and Development. (2009) Part 2: Annexes to the Detailed Review Paper on Transgenic Rodent Mutation Assays, Series on Testing and Assessment, N° 103. ENV/ JM/MONO(2009)29. OECD, Paris. Available at: http://www.oecd. org/officialdocuments/publicdisplaydocumentpdf/?cote=env/jm/ mono(2009)29doclanguage=en (accessed 21 January 2015). 41. Organisation for Economic Cooperation and Development. (2011) Trans- genic Rodent Somatic and Germ Cell Gene Mutation Assay: Retrospective Performance Assessment, Series on Testing and Assessment, N° 145. ENV/ JM/MONO(2011)20. OECD, Paris. Available at: http://www.oecd.org/ env/ehs/testing/48532170.pdf (accessed 21 January 2015). 42. United States Food and Drug Administration. (2006) Guidance for indus- try and review staff: recommended approaches to integration of genetic toxicology study results. Available at: www.fda.gov/downloads/Drugs/ GuidanceComplianceRegulatoryInformation/Guidances/ucm079257.pdf (accessed 21 January 2015). 43. Bemis, J.C, Labash, C., Avlasevich, S.L., Carlson, K., Berg, A., Torous, D.K., Barragato, M., MacGregor, J.T., and Dertinger, S.D. (2015) Rat Pig-a muta- tion assay responds to the genotoxic carcinogen ethyl carbamate but not the non-genotoxic carcinogen methyl carbamate. Mutagen. 30, 343–347. doi:10.1093/mutage/geu084. 44. Miura, D., Dobrovolsky, V. N., Kimoto, T., Kasahara, Y. and Heflich, R. H. (2009) Accumulation and persistence of Pig-A mutant peripheral red blood cells following treatment of rats with single and split doses of N-ethyl-N-nitrosourea. Mutat. Res., 677, 86–92. 45. Dertinger, S. D., Phonethepswath, S., Avlasevich, S. L. et al. (2012) Effi- cient monitoring of in vivo pig-a gene mutation and chromosomal dam- age: summary of 7 published studies and results from 11 new reference compounds. Toxicol. Sci., 130, 328–348. 46. Heddle, J. A., Dean, S., Nohmi, T. et al. (2000) In vivo transgenic mutation assays. Environ. Mol. Mutagen., 35, 253–259. 47. Thybaud, V., Dean, S., Nohmi, T. et al. (2003) In vivo transgenic mutation assays. Mutat. Res., 540, 141–151. 48. Organisation of Economic Co-operation and Development. (2013) Guideline for Testing of Chemicals: Test Guideline 488 - Transgenic Rodent Somatic and Germ Cell Gene Mutation Assays, adopted 26 July 2013, OECD, Paris. Available at: http://www.oecd-ilibrary.org/docserver/ download/9713251e.pdf (accessed 21 January 2015). 342 L. F. Stankowski et al., 2015, Vol. 30, No. 3 byguestonJanuary18,2017http://mutage.oxfordjournals.org/Downloadedfrom