You are on page 1of 12

Mutation Research 455 (2000) 155–166

In vivo rodent micronucleus assay:


protocol, conduct and data interpretation
Gopala Krishna a,∗ , Makoto Hayashi b
a Department of Worldwide Preclinical Safety, Parke-Davis Pharmaceutical Research,
Division of Warner-Lambert Company, 2800 Plymouth Road, Ann Arbor, MI 48105, USA
b Division of Genetics and Mutagenesis, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan

Abstract
In vivo rodent micronucleus assay has been widely used to detect genotoxicity. Evaluation of micronucleus induction is
the primary in vivo test in a battery of genotoxicity tests and is recommended by the regulatory agencies around the globe
to be conducted as part of product safety assessment. The assay, when performed appropriately, detects both clastogenicity
and aneugenicity. Methods for performing micronucleus evaluation have evolved since its initial description in the 1970s.
In recent years, the focus has been directed toward improving micronucleus detection with high efficiency by proposing
data-based recommendations to the standard initial protocol design. Such improvements include, e.g., the use of appropriate
harvest time(s), inclusion of one or both sexes, number of doses tested, limit dose, integrating micronucleus assessment into the
routine toxicology studies, use of fluorescent staining, automation of micronucleus detection and assessment of micronuclei in
multiple tissues. This protocol paper describes: the mechanism of micronucleus formation, a generalized protocol for manual
detection, enumeration of micronuclei, and data interpretation in light of published information thus far, on the regulatory
aspects of this assay. Certain recent protocol issues that are practical in nature are equally valid in relation to standard manual
method and provide robust database, which are also included for consideration. It is expected that such improvements of the
protocol will continue to drive the utility of this assay in the product safety assessment. © 2000 Elsevier Science B.V. All
rights reserved.
Keywords: Micronucleus assay; Rodents; Mice; Rats; Protocol; Automation; Recent methods; Integration with toxicology studies

1. Introduction assess the effects of chemicals on genetic mechanisms


and the consequent risk to organisms, including hu-
“The present generation is only a caretaker of the mans. Of equal importance are studies of the mecha-
human genome of future generations” — a statement nisms by which adverse genetic effects are mediated
made by Malling and Valcovic and quoted by Brusick and epidemiological studies of the frequency of ge-
[1] describes precisely the ultimate objective of ge- netic effects relative to chemical exposure. Thus far,
netic toxicologists. Genetic toxicology is the study of it is clear that information on three levels of mutation,
adverse effects on the process of heredity. Studies of e.g., gene, chromosomal, and cellular apparatus neces-
genetic toxicology have given rise to a number of test- sary for chromosome segregation, is necessary to pro-
ing procedures, both in vitro and in vivo, designed to vide broad coverage of the mutagenic and presumably
carcinogenic potential of a chemical or radiation. In
∗ Corresponding author. Tel.: +1-734-622-7985. this regard, micronucleus assay has been widely used
E-mail address: gopala.krishna@wl.com (G. Krishna). to measure genotoxicity, both in vitro and in vivo. The

0027-5107/00/$ – see front matter © 2000 Elsevier Science B.V. All rights reserved.
PII: S 0 0 2 7 - 5 1 0 7 ( 0 0 ) 0 0 1 1 7 - 2
156 G. Krishna, M. Hayashi / Mutation Research 455 (2000) 155–166

description of in vivo micronucleus assay is the pri- Visualization of micronuclei is facilitated in these
mary focus of this paper. The in vivo test is especially cells because they lack a main nucleus. An increase
relevant to assessing genotoxicity hazard in that it al- in the frequency of micronucleated PCE (MNPCE) in
lows consideration of factors of in vivo metabolism, test agent-treated animals is an indication of induced
pharmacokinetics, and DNA-repair processes and is chromosome damage. The PCEs, with time, lose
also useful in further investigation of a mutagenic ef- RNA and contain primarily hemoglobin and become
fect detected by an in vitro genotoxicity test. normochromatic erythrocytes (NCEs, mature erythro-
Evaluation of micronucleus frequency in vivo is the cytes [red blood cells], somewhat smaller than PCE,
primary test in a battery of genotoxicity tests and is acidophilic and stain light orange or orange-pink with
recommended by the regulatory agencies around the Giemsa). These two types of erythrocytes, which stain
globe to be conducted as part of product safety assess- differentially, can be seen in bone marrow, spleen,
ment. The assay, when performed appropriately, de- and blood compartments. Examples of micronucle-
tects both clastogenicity (chromosome breakage) and ated cells stained with Wright’s Giemsa method and
aneugenicity (chromosome lagging due to dysfunc- a column fractionation method that eliminates all
tion of mitotic apparatus). Micronuclei, also hemato- nucleated cells are shown in Figs. 2 and 3.
logically known as Howell–Jolly bodies, are generally In the micronucleus assay, the PCE-to-NCE ratio
smooth, round remnants of nuclear chromatin seen in between test agent-treated animals and vehicle-control
erythrocytes and are not to be confused for Heinz or animals provides a cytotoxicity index. During later
Heinz Ehrlich bodies resulting from oxidative injury stages of maturation, and on a needed basis, these
to and precipitation of hemoglobin. In vivo, the pro- cells move into the peripheral blood compartment.
cess of erythropoiesis (production of erythrocytes) has A kinetochore-labeling procedure [2] can distin-
been exploited in the micronucleus test. guish the mechanism of either a clastogen-induced
damage (primarily chromosome breakage and ab-
sence of kinetochore(s) in the micronucleus) or
2. Process of erythropoiesis an aneugen-induced spindle dysfunction (primarily
lagging chromosome(s) and the presence of kine-
The process of erythropoiesis and the mechanism of tochore(s) in the micronucleus). Examples of such
micronucleus formation in vivo are shown in Fig. 1. differentiation and aneuploidy evaluation are shown
In the adult rodent, both bone marrow and spleen in Fig. 4. However, other methods, e.g., DNA-specific
are hemopoietic organs, in which stem cells form the centromere labeling [3], can also be used to evaluate
basis of erythropoiesis with proliferation and matu- aneuploidy.
ration stages. During proliferation, the cells continue The micronucleus assay [4] is devised primarily for
to divide at which time a given test agent admin- evaluating the ability of test agents to induce structural
istered may act and cause chromosome damage, and/or numerical chromosomal damage. Both kinds of
such as breaks and exchanges, and may also act on damage are associated with the appearance and/or pro-
macromolecules related to the function of chromatid gression of tumors, and with adverse reproductive and
disjunction, e.g., tubulin causing spindle dysfunction, developmental outcomes. In most testing situations,
depending on the mechanism of action. These anoma- the test agent is administered acutely (generally once)
lies (a fragment or a whole chromosome) may lag to mice or rats, and the frequency of MNPCE deter-
behind in the cell during division and may not become mined in slides prepared from bone marrow harvested
integrated into daughter nuclei, rather may eventually 24 and 48 h after treatment. In some experimental sit-
form micronuclei, which can be seen in the cytoplasm. uations, peripheral blood can be sampled instead of
During maturation, when an erythroblast develops into bone-marrow and mature erythrocytes (NCE) scored
a polychromatic erythrocyte (PCE, young erythrocyte in addition to or instead of PCE for the presence of mi-
still contains RNA, is basophilic and stains light blue cronuclei. This assay has several important advantages
or blue gray with Giemsa), the main nucleus is ex- over the analysis of bone-marrow metaphase analy-
truded; any micronucleus that has been formed may sis. For example, it is technically simple, the endpoint
remain behind in the otherwise enucleated cytoplasm. scored is more objective and amenable for automa-
G. Krishna, M. Hayashi / Mutation Research 455 (2000) 155–166 157

Fig. 1. (a) The process of erythropoiesis in vivo; (b) the mechanism of micronucleus formation in the polychromatic erythrocytes (PCEs)
and normochromatic erythrocytes (NCEs). Also, classification of kinetochore-positive (K+) and kinetochore-negative (K−) erythrocytes.
N, nucleus; PEB, proerythroblast; MN, micronucleus.
158 G. Krishna, M. Hayashi / Mutation Research 455 (2000) 155–166

Fig. 2. A photomicrograph of rat whole bone-marrow smear showing nucleated as well as enucleated cells. The enucleated cells (PCEs
and NCEs) also contain micronuclei.

tion, it is less time consuming, the assay can detect such information is rarely used for safety assessment
both clastogens and aneugens, and it can be easily in- of test chemical along with the frequency of cells with
tegrated into general toxicology studies. Although the chromosome aberrations.
chromosomal aberration assay does allow for an as- A number of major methodological issues for the
sessment of the types of structural damage; however, in vivo rodent micronucleus assay were agreed upon

Fig. 3. A photomicrograph of column-fractionated rat bone marrow and cytospun slide preparation showing only enucleated cells and 2
PCEs containing micronuclei.
G. Krishna, M. Hayashi / Mutation Research 455 (2000) 155–166 159

Fig. 4. Photomicrographs of cells stained with antikinetochore antibody, fluoresceinated antibodies, and propidium iodide. (A) A metaphase
cell showing centromere specific staining of CREST serum (V79 Chinese hamster lung cell chromosomes, for example). (B) A microscopic
field showing cellulose-column fractionated cytospun cyclophosphamide-treated (clastogen) mouse bone-marrow PCE and NCE differen-
tiation; PCEs with kinetochore-negative micronuclei (arrows). (C) A microscopic field showing vincristine-treated (aneugen) spleen PCEs
with kinetochore-positive micronuclei (arrows).

at the International Workshop on Genotoxicity Test nical Requirements for Registration of Pharmaceuti-
Procedures (IWGTP), in Melbourne during 1993 [5]. cals for Human Use (ICH) [6] and also the revision of
This document provided guidance in formulating the OECD guidelines for testing of chemicals (NO. 474,
International Conferences on Harmonization of Tech- Mammalian Erythrocyte Micronucleus Test) [7]. As
160 G. Krishna, M. Hayashi / Mutation Research 455 (2000) 155–166

a follow-up and complementary to these, at the sec- 3.1.3. Test animal selection, identification, and
ond IWGTP held in Washington, DC, March 25–26, housing
1999, the expert working group discussed a number of Test animals: [mice or rats; Age: - - -; Weight: - - -
issues, including integration of the repeated-dose mi- Strain: - - -; Source: - - -]. Rationale for selection of
cronucleus assay into general toxicology studies [8], a test animal: [e.g., rats/mice are used as an animal
the use of automated scoring techniques, micronu- model to detect micronucleus induction because sub-
cleus evaluation in tissues other than bone marrow or stantial historical information is available and this
peripheral blood, and methods for differentiating mi- model has been utilized in other studies to obtain
cronucleus derived from acentric chromosome frag- toxicology information regarding the test agent under
ments and from centromeric chromosomes [9]. The study]. Generally, commonly used laboratory strains
reader of this protocol paper is strongly advised to re- of young healthy sexually matured animals, where
fer to these prior references for additional background bone marrow is expected to be actively dividing, are
detail and special circumstances in conducting and re- utilized and are acclimated to the laboratory envi-
porting micronucleus assay. ronment for a minimum 5 days and examined prior
to initiation of the study to ensure that they appear
healthy. Animals are assigned to study groups using
3. Acute micronucleus assay protocol a randomization method and necessary information is
listed for animal identification purposes using, e.g.,
cage cards and/or electronic implanted transponder
An example of a good laboratory practice (GLP)
chips. Animals are housed individually or in treat-
laboratory protocol using rodent bone marrow, with
ment groups of same sex in appropriate cages and
single dose and two harvest times is briefly described
appropriate laboratory-specific animal husbandry
here. Any variation of this protocol could be used,
procedures are followed, e.g., appropriate room tem-
depending on the objective, need, or level of interest
perature, humidity, light cycle, laboratory rodent diet,
by the researcher.
and unlimited supply of water.

3.1. Study no. and title: study 123: rodent 3.1.4. Test agent and controls
micronucleus assay of test agent(s) Test agent-specific number or name designation,
chemical name, therapeutic and/or chemical class (if
3.1.1. Assay principle any), source, active moiety, stability and homogeneity
Rodents are treated with the test agent by appropri- in vehicle may be included. Special storage and han-
ate route, bone marrow extracted at appropriate times dling conditions, assays for confirmation of potency
after treatment, smear slides are prepared either with following completion of treatment. Information on ve-
whole bone marrow or cellulose column-fractionated hicle (solvent control, generally nontoxic at the dose
cell suspension, stained, coded, and analyzed for the volume used and not known to produce chemical re-
toxicity (PCE to NCE ratio) and micronucleated cell action with the test agent, e.g., water, or methylcellu-
frequency. lose aqueous solution) and positive control [an agent
that is known to induce micronuclei, used to veri-
3.1.2. Purpose fy performance of the assay, e.g., cyclophosphamide
The purpose of the micronucleus assay is to identify i.p. −20 mg/kg for rats, 40 mg/kg for mice; mitomycin
test substance(s) that cause micronuclei formation as C i.p. −0.5 mg/kg rats and mice].
a result of lagging of chromosome fragments (clasto-
genicity) or whole chromosomes (aneugenicity), gen- 3.1.5. Route of administration and sample
erally in rodent bone-marrow erythropoietic cells. If preparation
the test agent is positive in the routine test, then, spe- Justification for route of administration (e.g., oral,
cific centromeric antibodies or DNA probes may be intraperitoneal, or intravenous) is generally included:
used to determine the mechanism of micronucleus for- this route has been intended for human exposure,
mation (e.g., clastogenic or aneugenic), if desired. and/or was used in a single-dose rat/mouse acute
G. Krishna, M. Hayashi / Mutation Research 455 (2000) 155–166 161

toxicity study of (test agent). Test agent dose calcula- Table of treatment groups
tions is based on active moiety, and given on a mg/kg
body weight basis at a dose volume 10 to 20 ml/kg. Treatment Dose Group number
The positive control, cyclophosphamide, e.g., is dis- (mg/kg)
solved in distilled water at 2 mg/ml and administered Sex Sex
at 20 mg/kg to rats or 40 mg/kg to mice by a single Vehicle control 0 M 1 (10)a F 6 (10)
i.p. injection of 10 ml/kg. Test agent Low M 2 (10) F 7 (10)
Test agent Mid M 3 (10) F 8 (10)
3.1.6. Test-agent dose selection criteria
Test agent High M 4 (10) F 9 (10)
Regulatory guidelines recommend that the high
Positive controlb 20/40 M 5 (5) F 10 (5)
dose selected for the rodent micronucleus assay should
produce some toxicity, be at maximum tolerated dose
a Values
in parentheses are the numbers of animals
(MTD), or be administered at 2000 mg/kg. Generally,
the MTD is the highest dose that can be adminis- dosed per group; b Positive control, e.g., cyclophos-
tered without inducing lethality or excessive toxicity phamide — 20 mg/kg for rats and 40 mg/kg for mice.
during the study causing moribund euthanasia. It has
also been recommended that the intermediate dose be However, if a test agent is relatively nontoxic, es-
one-half of the high dose and the low dose be one-half pecially at ≥2000 mg/kg and genotoxicity would not
of the intermediate dose. Information on acute toxi- be expected based on data from structurally related
cology data is included, if available or a dose-range agents, a full study using three dose levels may not
finding study matching schedule of treatment and/or be considered necessary and a limit dose test at 2000
euthanasia times used in the micronucleus assay may mg/kg may be sufficient.
be conducted to select appropriate doses.
3.1.8. Test procedures: animal observations,
3.1.7. Treatment groups, number and sex of animals euthanasia and bone-marrow processing
For a standard study, based on a regulatory recom- Animals are observed for clinical signs of toxicity at
mendation regarding the in vivo micronucleus assay, various intervals after treatment, and at 24 and 48 h or
a single dosing regimen and two euthanasia times (24 at the discretion of study scientist. Bone-marrow cells
and 48 h) were selected for this study. If the test agent will be harvested, cells processed and/or slides pre-
does cause differential toxicity in males and females, pared for evaluation according to standard procedures.
then use of both sexes is necessary, otherwise the Briefly, animals will be euthanized by CO2 asphyxi-
use of an appropriate sex, usually male, is sufficient. ation and one usable femur or tibia excised, skin and
In the case where sex-related difference in toxicity muscle tissue trimmed, and both ends of the bone tips
is apparent, groups of five males and five females severed with bone snips, marrow flushed gently from
scheduled for the 24- and 48-h euthanasia will be the channel into a tube with fetal bovine serum (FBS,
administered vehicle control. Generally, three dose approximately 3 ml/femur). Cells are processed for
levels of test agent are used and each treatment group manual (standard smear or cellulose column fraction-
consists of five animals per sex per euthanasia time. ation, Appendix A [10,11] or flow cytometry method
If unexpected lethality is anticipated at the high dose, of evaluation of micronuclei according to procedures
a few extra animals may be included at this group to described in the literature [9,12–14].
replace, if necessary. Also, additional animals may
be included at each group, if exposure (plasma drug 3.1.9. Data collection, analysis, and evaluation and
concentration) data are needed as part of the micronu- interpretation
cleus assay. Positive control animals are included In the micronucleus assay, the proportion of MN-
only at the 24-h euthanasia time. Five animals of each PCE constitute the primary endpoint and the pro-
sex per group will be euthanized in a sequence at 24 portion of PCEs is the supportive endpoint to assess
and 48 h after treatment and bone marrow harvested cytotoxicity, which helps demonstrate a target cell ex-
according to the following schedule: posure with the test chemical. Generally, in the manual
162 G. Krishna, M. Hayashi / Mutation Research 455 (2000) 155–166

method, the slides are coded to not to reveal the treat- 3.1.10. Test report procedure
ment groups by the scorer and 2000 PCE/animal are At the conclusion of the study, the results of the
evaluated for the presence of micronuclei. The unit of study are reported in a report, with a study number and
analysis is PCE and not the number of micronuclei per a report number, giving the experimental design, pro-
PCE, as a PCE may contain more than one micronu- tocol, raw data, evaluation of results, and a conclusion.
cleus. For bone-marrow toxicity, at least 200 total ery- Archival and retrieval information is also included in
throcytes are evaluated per animal for the proportion the report. An example of information typically in-
of PCE in bone marrow and 1000 in peripheral blood. cluded in a research report is shown in Appendix B.
In the automated method (e.g., flow cytometry), the
number of cells evaluated could be much higher and 3.1.11. Standard operating procedures, GLP
up to 100,000 total erythrocytes and the proportions compliance, laboratory safety, and study schedule
of PCE, NCE, MNPCE and MNNCE may be quan- The methods used in the study are described in de-
tified depending on the purpose of the study. Indivi- tail in the laboratory-specific Manual of Operations. A
dual animal data are listed in a tabular form along with statement as to whether the study will be conducted in
means, standard deviations and statistical significance, compliance with the OECD or any other global regu-
if any. latory body’s GLP regulations for nonclinical labora-
Numerical data are analyzed using appropriate sta- tory studies, is included. Exceptions to this may be
tistical tests, e.g., MNPCE data are analyzed with a stated, as needed in exploratory studies. A statement
one-sided test for an increase and PCE data may be as to the potential toxicity of positive control (e.g.,
also be analyzed with a one-sided test for a decrease. cyclophosphamide), is included and appropriate pre-
If there is no evidence for a difference in response cautions taken to minimize exposure, as well as steps
between sexes, the data from both sexes may be com- to be taken if exposure takes place, by accident, are
bined for statistical analysis. included. Information regarding tentative start date,
While evaluating data, both statistical and biologi- completion date, and final report date are included
cal criteria are considered. One criterion for a positive prior to protocol approval by the study director and
result would be a statistically significant dose-related the facility management.
increase in MNPCE frequency at any time point
with at least 1 value significantly exceeding the his-
torical vehicle control range. Equivocal results may 4. Repeat-dose integrated micronucleus study
be clarified by further testing using, preferably, the
modified experimental conditions. Positive results In the industrial toxicology laboratories, micronu-
indicate that the test agent induces micronuclei un- cleus assay can be integrated into routine toxicologi-
der the experimental conditions, which have been cal studies [8]. This approach utilizes: (i) the general
the result of chromosome damage and/or damage principles of toxicology that govern the overall toxi-
to the mitotic apparatus. Negative results mean that city profile of a test substance; (ii) factors, such as
the test agent does not produce micronuclei under the dose and/or route of administration, metabolism,
the experimental conditions. The data may be put in principles of toxicokinetics, and saturation of de-
perspective by taking test agent’s pharmacokinetic fense mechanisms, are considered in evaluating
and metabolism pathways and systemic toxicity into genotoxicity; (iii) the concept of administering mul-
consideration. If a positive micronucleus response for tiple tolerable doses achieving steady-state plasma
the test agent is obtained, additional studies can be drug concentrations that are more relevant in human
conducted to identify the mechanistic origin of the risk assessment compared to high acute doses; and
micronuclei. Generally, micronuclei that arise from (iv) this approach would minimize the amount of test
clastogenic damage lack a kinetochore, and those compound, number of animals, and other resources
associated with numerical chromosomal damage con- that are generally utilized in the conduct of standard
tain a kinetochore. The presence of a kinetochore can acute micronucleus assay.
be identified using commercially available antibodies While integrating micronucleus assessment into
[2,3]. general toxicology repeat-dose studies, e.g., 2-to-4-
G. Krishna, M. Hayashi / Mutation Research 455 (2000) 155–166 163

Table 1
Rodent micronucleus assay: historical control database (1987–1998): Parke-Davis Pharmaceutical Laboratory
PCEa /100 TEb MNPCEc /1000 PCE

Males Females M+F d Males Females M+F

Mice
Vehicle control
No. of mice 243 245 430 230 215 430
No. of assays 47 45 44 47 45 44
Mean assay range 42.0–66.9 48.1–75.6 46.0–69.7 0.4–3.8 0.6–3.6 0.9–3.1
Positive control (CPe - 40 mg/kg)
No. of mice 130 130 230 115 115 220
No. of assays 24 24 23 24 24 23
Mean assay range 42.9–67.6 39.6–69.3 41.3–66.8 7.7–42.7 8.0–44.7 8.8–42.1
Rats
Vehicle control
No. of rats 185 180 360 185 180 360
No. of assays 37 36 36 37 36 36
Mean assay range 40.1–63.3 34.0–65.0 37.1–59.6 1.1–6.4 0.8–4.9 1.3–5.3
Positive control (CP - 20 mg/kg)
No. of rats 135 120 240 135 120 240
No. of assays 24 24 24 24 24 24
Mean assay range 25.5–54.9 25.3–50.8 27.8–52.7 9.0–43.8 5.8–25.1 10.4–33.8
a Polychromatic erythrocytes.
b Totalerythrocytes.
c Micronucleated polychromatic erythrocytes.
d Combined sexes, where available.
e Cyclophosphamide. Data based on CD1 mice and Wistar rats of age 5 to 8 weeks at study initiation.

week studies and possibly 13-week studies, the fol- from a study, the routine use of positive control in
lowing approach may be followed. Generally, a femur every micronucleus assay has been questioned by the
or tibia from five animals/sex/group used for the scientific community, especially in laboratories which
toxicological assessment is utilized for micronucleus have demonstrated assay reproducibility and conduct
assay. Positive control animals may be included to studies under GLP regulations. Based on a review
match with the euthanasia schedule of study animals of available data, such as shown in Table 1, on the
and given a single 20-mg/kg i.p. dose of cyclophos- reproducibility of positive control response, the mi-
phamide dissolved in sterile water at a concentration cronucleus assay Expert Panel recently recommended
of 2 mg/ml and administered i.p. at a dose volume of that the use of positive control may not be necessary
10 ml/kg for rats. Body weights of the positive con- in every study [9].
trol animals are determined prior to dosing and any
clinical signs noted at termination may be recorded.
Appendix A. Micronucleus assay
The positive control animals are treated and bone
marrow collected approximately 24 h after treatment. A.1. Preparation of cells and/or slides
Inclusion of both vehicle and positive controls is in-
tended to minimize any staining variations in slide A.1.1. Standard smear method with Wright’s Giemsa
evaluation and/or to adjust automated measurement The bone marrow with serum in tubes is centrifuged
settings, if appropriate. The data are collected similar at approximately 150×g for 5 min, the supernatant re-
to acute micronucleus assay. In light of minimizing moved, the pellet resuspended using a few drops of ad-
animal use in research and still obtain required data ditional serum, if necessary. A drop of cell suspension
164 G. Krishna, M. Hayashi / Mutation Research 455 (2000) 155–166

is placed onto the slide and using a pusher slide a smear fully, dropped into the center of the column so that it
is prepared. Generally at least two slides are prepared is absorbed outwards to the plastic edge of the cellu-
per animal. Slides are air dried, fixed in methanol, lose column.
and stained with 5% to 10% Wright’s Giemsa with Following this, 20 ml Hanks’ balanced salt solu-
Haemastainer. All slides are cover-glassed using Per- tion (HBSS, without phenol red) is carefully added to
mount and coded for evaluation. the column surface and approximately 20 ml eluted
(takes 20 to 30 min). The eluate generally contains ery-
A.1.2. Cellulose column fractionation method throcytes. The eluate is centrifuged at approximately
Slides may also be prepared according to the cel- 800×g for 10 pellet resuspended in a few drops of FBS
lulose column fractionation and cytocentrifugation for smears, or resuspend the pellet in 200 ␮l of sup-
[10,11]. The most common method of preparing plemented MEM culture media for cytospun smears.
slides is to use whole bone marrow, which contains For manual micronucleus scoring, slides are prepared
various types of blood cells and their precursors. A using a cytocentrifuge and labeled sufficiently to iden-
cellulose column fractionation procedure has been tify the treatment group and experiment.
recommended to remove all nucleated cells as well Slides are stained for micronucleus analysis
as artifact-producing cell debris from bone marrow in a routine fashion with Wright’s Giemsa tech-
to facilitate scoring of micronuclei [10,11]. With nique using a Haemastain automatic staining ma-
this technique, one can overcome the problem of chine, coverslipped, blind coded, and used for data
micronucleus-imitating mast cell granules, espe- collection.
cially, in rat bone marrow. The column-fractionated Micronuclei are identified according to the criteria
cells may be used for making standard smears, for established by Schmid [4] and are darkly stained (pur-
cytocentrifugation, or for flow cytometric analysis ple) and generally round or almond shaped, although
depending on the objective of the experiment. If a lightly stained, ring shaped micronuclei occasionally
column method is used, each laboratory should assure occur. Micronuclei have sharp borders and are gen-
that micronucleated cell frequencies are obtained, erally 5–20% the size of the PCE and may occur
which are comparable with those obtained using di- in either PCE or NCE. However, in a single-dose
rect scoring. For this purpose, samples should be regimen only MNPCEs are counted. The unit of scor-
used that contain elevated micronucleus frequencies ing is the MNPCE, not the number of micronuclei
induced by a known clastogen and also an aneugen per PCE, as occasionally more than one micronu-
[9]. cleus may appear per PCE. In a repeated dosing
regimen, in addition to the bone-marrow analysis,
A.1.3. Preparation of cellulose column [10,11] micronuclei may be scored in PCEs and/or NCEs
One part of microcrystalline cellulose and one part found in the blood at the discretion of the study
of ␣-cellulose fiber are weighed, mixed by shaking in scientist. Initially, the ratio of PCE to total erythro-
a securely capped bottle for approximately 3 min. A cytes (PCE+NCE) for each animal is determined
20-mm disc of microscope-cleaning tissue is placed by examining at least 200 and 1000 erythrocytes for
at the bottom of a 20 ml plastic syringe and 1.0 to bone-marrow and peripheral blood, respectively, per
1.1 g of cellulose mixture is added to the column, animal and then, the number of MNPCE in 2000
packed by tapping the syringe on a hard surface (ap- PCE/animal is determined and data recorded on the
proximately 20 sharp taps from a height of about appropriate assay form and data are uncoded by the
1 cm), followed by slightly pressing down the cel- study scientist and analyzed by appropriate statistical
lulose mixture with a modified syringe plunger un- tests.
til the 3 ml mark of the syringe is reached. Bone
marrow is isolated in a routine fashion using 3 ml A.1.4. Slide staining using acridine orange [15]
FBS per two femurs in mouse and 3 ml FBS per The acridine orange stock solution is prepared as a
one femur in rat. Bone marrow is aspirated and dis- 0.1% aqueous solution that could be available for sev-
charged about 20 times using Pasteur pipette to break eral weeks stored at 4◦ C. Acridine orange, 0.24 mM
up clumps. Using a pipette, cell suspension is care- in 1/15 M Sörensen’s phosphate buffer (pH 6.8) (two
G. Krishna, M. Hayashi / Mutation Research 455 (2000) 155–166 165

parts of stock solution and 30 parts of the buffer), is B.4. Test conditions
used as a working solution. The fixed cells are stained
in this solution for 3 min at room temperature. The Positive and negative control data, data from
slides are rinsed in the buffer three times for 1–3 min range-finding study, if conducted, rationale for dose
each time. If the nuclei emit a reddish fluorescence, the selection, details of test agent preparation, details
slides are rinsed for another several minutes to opti- of the administration of the test agent, rationale for
mally stain nuclei with green fluorescence. The prepa- route of administration, methods for verifying that
rations are mounted with the same buffer, and sealed the test agent reached the general circulation or target
with Balsam paraffin or suitable media. An alternative tissue, if applicable, details of food and water quali-
more simple method can be used: one drop of 0.04 ty, detailed description of treatment and sampling
mM acridine orange solution in the same Sörensen’s schedules, methods of slide preparation, methods for
phosphate buffer is placed on the fixed cells and cov- measurement of toxicity, criteria for scoring MNPCE,
ered with cover slip. The excess solution is blotted and number of cells analyzed per animal, criteria for con-
sealed if necessary. The slide is already ready for fluo- sidering studies as positive, negative, or equivocal.
rescent microscopy. Observations can be made within
a day using fluorescent microscopy equipped with blue B.5. Results and discussion
excitation and 515–530 nm barrier filter. Cytoplasm of
PCE emits red fluorescence and micronuclei as well Clinical signs of animal toxicity, percentage of
as nucleus of nucleated cells fluoresce yellowish green PCE among total erythrocytes or ratio of PCE to
or yellow. NCE, number of MNPCE, given separately for each
animal, mean±standard deviation of MNPCE per
group, treatment–response relationship, where possi-
Appendix B. Test report ble, statistical analyses and methods applied, concur-
rent and historical negative-control data, concurrent
The research report should include the following and historical positive control data, where applicable,
information. and discussion of the results, as appropriate.

B.6. Conclusion
B.1. Test agent
A one or two sentence conclusion as to whether or
Source, identification data and CAS No., if known, not the test agent was negative, positive, or equivocal
physical nature and purity, physiochemical properties under the conditions of the assay.
relevant to the conduct of the study, stability of the
test agent, if known. B.7. Authorized signatures and GLP compliance
statement (if necessary)
B.2. Solvent/vehicle
Signatures of study director, responsible staff and
Justification for choice of solvent/vehicle; solubi- management, are included. A GLP compliance state-
lity, homogeneity, and stability of the test agent in the ment along with quality assurance unit’s statement is
solvent/vehicle, if known. also included.

B.3. Test animals References

Species/strain used, number, age, and sex of ani- [1] D. Brusick, Principles of Genetic Toxicology, 2nd edn.,
mals, source, housing conditions, diet, individual Plenum, New York, 1987.
[2] G. Krishna, R. Fiedler, J.C. Theiss, Simultaneous evaluation
weight of the animals at the start of the test, including of clastogenicity, aneugenicity, and toxicity in the mouse
body weight range, mean, and standard deviation for micronucleus assay using immunofluorescence, Mutat. Res.
each group. 282 (1992) 159–304.
166 G. Krishna, M. Hayashi / Mutation Research 455 (2000) 155–166

[3] B.M. Miller, H.F. Zitzelsberger, H.U. Weier, I.-D. Adler, A. Russo, S. Sutou, In vivo rodent erythrocyte micronucleus
Classification of micronuclei in mouse erythrocytes: assay: II. Some aspects of protocol design including
immunofluorescent staining using CREST antibodies repeated treatments, integration with toxicity testing, and
compared to in situ hybridization with biotinylated gamma automated scoring, Env. Mol. Mutagen. 35 (2000) 234–
satellite DNA, Mutagenesis 6 (1991) 297–302. 252.
[4] W. Schmid, The micronucleus test, Mutat. Res. 31 (1975) [10] F. Romagna, C.D. Staniforth, The automated bone marrow
9–15. micronucleus test, Mutat. Res. 213 (1989) 91–104.
[5] M. Hayashi, R.R. Tice, J.T. MacGregor, D. Anderson, D.H. [11] G. Krishna, J. Theiss, Concurrent analysis of cytogenetic
Blakey, M. Kirsch-Volders, F.B. Oleson Jr., F. Pacchierotti, F. damage in vivo: a multiple endpoint-multiple tissue approach,
Romagna, H. Shimada, S. Sutou, B. Vannier, In vivo rodent Environ. Mol. Mutagen. 25 (1995) 314–320.
erythrocyte micronucleus assay, Mutat. Res. 312 (1994) 293– [12] K. Criswell, G. Krishna, D. Zielinski, G. Urda, J. Theiss,
304. P. Juneau, M. Bleavins, Use of acridine orange in: flow
[6] L. Muller, Y. Kikuchi, G. Probst, L. Schechtman, H. cytometric evaluation of erythropoietic cytotoxicity, Mutat.
Shimada, T. Sofuni, D. Tweats, ICH-harmonized guidances on Res. 414 (1998) 49–61.
genotoxicity testing of pharmaceuticals: evolution, reasoning [13] K. Criswell, G. Krishna, D. Zielinski, G. Urda, J. Theiss,
and impact, Mutat. Res. 436 (1999) 195–225. P. Juneau, M. Bleavins, Use of acridine orange in: flow
[7] OECD, 1997, Guideline for the testing of chemicals. cytometric evaluation of micronuclei induction, Mutat. Res.
mammalian erythrocyte micronucleus test. Guideline 474, 414 (1998) 63–75.
July, 1997. [14] G. Krishna, K. Criswell, D. Zielinski, G. Urda, P. Juneau,
[8] G. Krishna, G. Urda, J. Theiss, Principles and practices of M. Bleavins, J. Theiss, Validation of micronucleus evaluation
integrating genotoxicity evaluation into routine toxicology in the rat using flow cytometry with five model compounds,
studies: a pharmaceutical industry perspective, Environ. Mol. Environ. Mol. Mutagen. 31 (1998) 46.
Mutagen. 32 (1998) 115–120. [15] M. Hayashi, T. Sofuni, M. Ishidate Jr., An application of
[9] M. Hayashi, J.T. MacGregor, D.G. Gatehouse, I. Adler, acridine orange fluorescent staining to the micronucleus test,
D.H. Blakey, S.D. Dertinger, G. Krishna, T. Morita, Mutat. Res. 120 (1983) 241–247.

You might also like