Sei sulla pagina 1di 24

Immune Cell Phenotyping Using Flow UNIT 18.

8
Cytometry
Immunophenotyping is a general term used to describe the use of fluorescently labeled
antibodies to identify and quantify distinct subpopulations of cells within a heteroge-
neous population of cells. Frequently, the term is used more specifically to refer to the
characterization of subsets of cells associated with the immune system (i.e., immune cell
phenotyping). In either case, the antibodies used for immunophenotyping are usually
specific for cell surface proteins that are known to be differentially expressed on discrete
subsets of cells. Immunophenotyping can be carried out using either flow cytometry or
immunohistochemical analysis. However, flow cytometry is often the method of choice,
because it permits the acquisition of nonsubjective data on thousands of cells within sec-
onds. Furthermore, each cell suspension can be labeled with several different antibodies
simultaneously, and the fluorescence associated with each antibody can be collected on
a per-cell basis as correlated data. Modern flow cytometers can routinely measure five
or more different fluorescence emissions simultaneously, providing the means to easily
detect unique subsets within a heterogeneous population of cells. More recently, cell
surface phenotyping with immunofluorescent probes has been performed to ascertain
the functional status (e.g., DNA or cytokine content, mitochondrial membrane integrity,
glutathione status) of cells, making multiparameter flow cytometry a very powerful tool
for characterizing the activation state of a cell. The only drawback of flow cytometry
is that cells must be stained and analyzed in a single-cell suspension, which is easier
to prepare from some tissues than from others. Because cells derived from blood and
lymphoid tissues are readily maintained in suspension, flow cytometry has been widely
used to phenotype these cells.

Immunophenotyping of lymphoid tissues, particularly spleen and thymus, has been used
in the field of immunotoxicology for many years as part of a tiered screening approach
for detecting immunotoxicity following chemical exposure. Since thymic involution fre-
quently occurs after toxicant exposure, changes in thymocyte immunophenotypes are
commonly evaluated. Similarly, just as white blood cell (WBC) counts and differentials
have been used for many years to assess immune status in toxicant-exposed animals,
newer screening approaches such as immunophenotyping of spleen and/or lymph nodes
have been used to quantify frequencies of the major lymphocyte subtypes (B cells, T cells,
and T cell subsets) following chemical exposure. In studies conducted by the National
Toxicology Program, the utility of immune cell phenotyping has been validated using a
large database of environmental toxins and certain pharmaceutical agents. In these studies,
changes in lymphocyte phenotypes in the spleen or thymus following toxicant exposure
was identified as one of the best single correlates with toxicant-induced changes in host
resistance to pathogens or tumors (Luster et al., 1993). However, even though immunophe-
notyping of the spleen and thymus may correlate with toxic chemical exposure, it is not
necessarily a robust predictor of immunotoxicity, particularly when naive animals are
exposed to toxicants. Some highly immunotoxic chemicals, including cyclosporin A and
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), do not alter splenic immunophenotypes in
the absence of antigen challenge (Kerkvliet and Brauner, 1990; Vandebriel et al., 1999).
Likewise, changes in thymic phenotypes can occur indirectly, through stress-induced ef-
fects on the thymus that are independent of direct immunotoxicity. Thus, it is generally
agreed that immunophenotypic analysis of B cells, T cells, and T cell subsets present in
the thymus and/or spleen should be used in conjunction with conventional assessments
of immune function when screening for the immunotoxic effects of drugs and chemicals
(Immunotoxicology Technical Committee, 2001; Food and Drug Administration, 2002).
Immunotoxicology
Contributed by Julie A. Oughton and Nancy I. Kerkvliet 18.8.1
Current Protocols in Toxicology (2005) 18.8.1-18.8.24
Copyright 
C 2005 by John Wiley & Sons, Inc.
Supplement 23
Although immunotoxicology studies have focused primarily on the phenotypic analysis
of B cells and T cell subsets in spleen and/or thymus from naive mice, several extended
applications of immunophenotyping are emerging in the immunotoxicological literature.
One important extension is the analysis of peripheral blood cells from mice and rats
(Oughton et al., 1995; Nygaard and Lovik, 2002; Funatake et al., 2004). The use of
peripheral blood samples from rodents allows more direct comparisons to be made with
toxicity data that may be available from human studies, which nearly always rely on
peripheral blood samples. Analysis of blood also allows evaluation of systemic changes
that may result from toxicant effects in lymphoid tissues, as well as the testing of multiple
samples obtained from the same animal over an extended time.

Another important addition to standard immunophenotyping techniques is the analysis


of activation markers. Most cells in the immune system respond to antigenic stimulation
by altering their expression of a number of different proteins on the cell surface. Many
of these proteins are known to be involved in the functional response of the cell (e.g.,
adhesion, migration, costimulation), while others may reflect the cell’s antigenic history
(as in memory cells). Evaluation of activation markers is likely to provide an enhanced
ability to detect immunotoxic effects. For example, when mice were exposed to low doses
of TCDD for more than a year, no changes in major T- and B cell subsets were observed.
However, analysis of the naive/memory phenotypes of CD4+ T cells revealed that TCDD
exposure had significantly reduced the proportion of memory cells present (Oughton
et al., 1995). Likewise, evaluation of activation markers in the context of an ongoing
immune response may provide novel insight into the selective targets and mechanisms of
action of immunotoxic chemicals. Several laboratories are now using immunophenotyp-
ing to track the effects of toxicants on the activation and fate of antigen-specific T cells
following antigenic challenge, with very exciting results (Shepherd et al., 2000; Mitchell
and Lawrence, 2003; Funatake et al., 2004).

Since the specific antibodies that one might use for immunophenotyping will vary de-
pending on the species tested, the flow cytometer used, and the experimental questions
being asked, the following protocol describes a generic approach to the staining of cells
with fluorescently labeled monoclonal antibodies (MAbs) to assess cell surface protein
expression. Work in the authors’ laboratory has primarily involved mouse cells; however,
all aspects of this unit easily apply to the analysis of cells from rats, dogs, and horses.
The first protocol (Basic Protocol 1) describes the procedure by which cells obtained
from lymphoid tissue are stained with fluorochrome-conjugated MAbs (direct staining
method). A second protocol (Alternate Protocol) describes two indirect methods for the
staining of cells with primary MAbs that are not conjugated to a fluorochrome. Finally,
a third protocol (Basic Protocol 2) describes the staining of peripheral blood cells with
MAbs.

Support protocols are presented for the preparation of lymphoid cells (Support Protocol 1),
the fixation of cells (Support Protocol 2), and discrimination of dead cells (Support
Protocol 3). When freshly stained cells cannot be analyzed immediately, samples must
be fixed so that the cells are stable for storage (Support Protocol 2). Furthermore, because
the inclusion of dead cells in flow cytometric analysis can lead to the misinterpretation
of data and possibly to the rendering of erroneous conclusions, it is desirable to assess
cell viability using DNA dyes (Support Protocol 3).

Finally, a procedure for the determination of optimal antibody concentrations is presented


in Support Protocol 4.
Immune Cell
Phenotyping NOTE: The investigator should consult the manufacturer’s instruction manual for specific
Using Flow information regarding the operation of his or her flow cytometer.
Cytometry

18.8.2
Supplement 23 Current Protocols in Toxicology
DIRECT STAINING OF CELLS PREPARED FROM LYMPHOID TISSUES BASIC
PROTOCOL 1
This is the preferred method for staining cells obtained from lymphoid tissues (e.g., spleen,
thymus, lymph nodes). Basic Protocol 1 is appropriate for immunophenotyping of cells
from either rats or mice. Because this method uses MAbs that are directly conjugated
to fluorochromes, staining is simple and quick and usually produces little background
signal.

Toxicology studies usually involve the assessment of lymphoid tissues from several in-
dividual animals in a number of different treatment groups. For example, a 3-dose-plus-
control experiment that examined spleen and thymus from 5 animals per treatment group
would yield 40 individual cell samples that needed to be stained, usually in more than
one staining configuration. Processing of large numbers of samples can be facilitated by
staining cells in V-bottom 96-well microtiter plates, as opposed to 12 × 75–mm tubes.
These plates can be centrifuged using a microtiter plate carrier, and the contents of the
wells can be mixed easily using a vortex mixer (such as the Vortex-Genie 2, VWR Scien-
tific Products) equipped with a platform head for a microtiter plate. Use of a multichannel
pipettor (such as the 12-channel Pipet-Lite multichannel pipet from Rainin Instrument)
facilitates rapid dispensing of media into the wells, as well as quick transfer of samples
to Titertubes (Bio-Rad) for flow cytometric analysis. Titertubes are small tubes that can
easily be inserted into 12 × 75–mm disposable tubes, which are commonly used to run
samples on a flow cytometer. The abovementioned products greatly increase productivity
and also simplify the processing of large numbers of samples. To help keep track of
stained samples, a template of the microtiter plate can be prepared in which descriptions
of the contents of each well (e.g., animal source, MAbs used) are recorded. In lieu of
microtiter plates, cells can be stained in 12 × 75–mm plastic tubes or microcentrifuge
tubes.

Materials
Tissue of interest (e.g., lymphoid cells, Support Protocol 1)
Fc receptor (FcR)-blocking immunoglobulin (Ig; e.g., normal rat IgG) solution,
200 µg/ml
MAbs or isotype Igs directly conjugated to fluorochromes
PBS supplemented with sodium azide and BSA (PAB; see recipe)
Micropipettor or multichannel pipettor with tips
96-well V-bottom microtiter plates, disposable 12 × 75–mm polystyrene tubes, or
microcentrifuge tubes
Centrifuge, refrigerated and equipped with a microtiter plate carrier
Vortex mixer equipped with a platform head for a microtiter plate
Titertubes (Bio-Rad Laboratories; optional)
Additional reagents and equipment for preparation of lymphocyte suspension to be
analyzed (Support Protocol 1)
NOTE: Several steps need to be performed to minimize antibody cross-linking and the
subsequent internalization of cell surface receptors that may occur when these receptors
are bound to MAbs. This process is often referred to as capping of the antigen-antibody
complex. Some cell surface molecules are less likely to cap than others; however, surface
immunoglobulin on B cells can cap almost instantaneously. The addition of sodium azide
(final concentration, 0.1% w/v) to the staining medium will help to prevent capping. In
addition, all cell preparations and staining procedures, including wash steps, should be
performed at 4◦ C, a condition achieved by incubating the cells on ice. Staining samples
on ice also reduces the off-rate constant associated with antibody binding by a factor of
10 compared with the off-rate constant at 25◦ C.
Immunotoxicology

18.8.3
Current Protocols in Toxicology Supplement 23
Prepare lymphocyte samples for staining
1. Prepare a single-cell suspension of lymphocytes in 5% HBSS. Lyse red blood cells
(RBCs) if present in large numbers (Support Protocol 1).
2. Using a micropipettor or microchannel pipettor, dispense 1–4 × 106 cells into each
well of a V-bottom 96-well microtiter plate.
Alternatively, dispense cells into 12 × 75–mm tubes or microcentrifuge tubes.

3. Pellet cells by centrifugation for 3 min at 200 to 1500 × g, 4◦ C.


The range of centrifugation speeds quoted in this step reflects the fact that no one speed is
accepted by the entire flow cytometry community. Although 1500 × g may sound excessive,
this is the speed most commonly used by Stewart and Stewart (2001a) in order to minimize
the loss of cells during washing.

4. Remove supernatant by holding the plate over a sink and inverting with a flick of the
wrist or by decanting liquid from tubes. Blot the plate or the lip of each plastic tube
on an absorbent towel to remove excess supernatant.
5. Vortex plate or tubes lightly to resuspend cell pellet.
Label with fluorochrome-conjugated MAbs
6. Add 50 µl FcR-blocking Ig (10 µg) to each cell suspension. Vortex cells lightly and
incubate on ice for 10 min.
A number of cells (especially myeloid cells) have receptors for the Fc portion of an
immunoglobulin and will bind to any MAb. In order to assess the antigen-specific binding
of a MAb, FcR must be blocked using normal IgG from the same animal species as the
selected MAb. For example, for cells stained with rat anti–mouse CD4 MAb, normal rat
IgG should be used to block FcR-mediated binding. As an alternative, a MAb specific for
mouse FcR (CD16/CD32) can be used, although this involves a substantially higher cost.
FcR should not be blocked when assessing FcR expression.

7. Without removing the FcR-blocking reagent, add the appropriate fluorochrome-


labeled MAbs (see Support Protocol 4 for determination of optimal antibody con-
centrations) or isotype Igs to each well or tube. Vortex plate or tubes and incubate
cells on ice for 10 min, protected from light.
Cells can be stained with a number of MAbs simultaneously as long as there are no
interactions among these MAbs. For large studies, a cocktail of all the MAbs of interest
can be used to stain each sample. For example, if cells are to be stained for CD3, CD4,
and CD8, a single cocktail including all three MAbs can be prepared in a total volume of
10 µl. Cocktail staining greatly expedites sample preparation and ensures that all samples
are stained uniformly.
Over time, MAbs in solution tend to form aggregates. Cells that express Fc receptors can
bind these antibody aggregates with far greater avidity than they can antibody monomers.
Aggregates should be removed by microcentrifugation of the antibody solution for 5 min
at 12,000 to 13,000 rpm, 4◦ C, prior to staining.
Fluorochromes [especially the phycoerythrin (PE)–Cy5 and PE-Cy7 tandem conjugates]
are particularly sensitive to photodegradation. Therefore, all staining procedures should
be conducted in subdued light.

8. Add 150 µl PAB to each well or tube and centrifuge for 3 min at 200 to 1500 × g,
4◦ C. Remove supernatant as in step 4.

Immune Cell
Phenotyping
Using Flow
Cytometry

18.8.4
Supplement 23 Current Protocols in Toxicology
9. To remove unbound MAb, which can contribute to background fluorescence, wash
cells once by adding PAB (total volume in microtiter plate not to exceed 200 µl)
to each well or tube and centrifuging for 3 min at 200 to 1500 × g, 4◦ C. Remove
supernatant as in step 4.
10. Resuspend cells in 500 µl PAB with gentle vortexing.
For samples prepared in microtiter plates, resuspend each cell pellet in 200 µl PAB. Next,
using a multichannel pipettor, transfer the contents of each well to a Titertube and add an
additional 300 µl PAB, for a total volume of 500 µl. These Titertubes can then be inserted
into 12 × 75–mm plastic tubes for flow cytometric analysis.

11. Keep samples on ice until analyzed using a flow cytometer.


If stained cells cannot be analyzed on the day of preparation, they must be fixed for
longer-term storage (Support Protocol 2).

INDIRECT STAINING OF CELLS PREPARED FROM LYMPHOID CELLS ALTERNATE


PROTOCOL
In general, it is best to stain lymphoid cells with a MAb that is directly conjugated to a
fluorochrome (i.e., using the direct method). It is relatively easy to obtain MAbs specific
for mouse or rat markers in a number of conjugated forms from commercial sources.
However, there are very few mouse or rat MAbs that are conjugated to PE–Texas Red.
This will impact studies requiring four or five colors for immunophenotypic analysis,
since one MAb may require a second-step staining reagent. This concern is even more
relevant when dealing with species other than mice, rats, or humans, as it is very difficult
to find commercial sources of fluorochrome-conjugated MAbs for such species. In those
situations, it may be necessary to resort to one of two indirect staining methods.

In one method, cells are incubated first with an unconjugated primary MAb and then with a
fluorochrome-conjugated antibody that is specific for the primary antibody. For instance,
if cells are stained with an unconjugated rat IgG2a MAb that is specific for mouse CD4,
the secondary antibody would be a fluorochrome-conjugated antibody that is specific
for rat IgG. In most cases, the secondary antibody will be a polyclonal anti-Ig antibody,
although Ig isotype–specific MAbs can sometimes be obtained from commercial sources.
In this example, one can limit cross-reactivity of the secondary antibody with all other IgG
isotypes by restricting the specificity of the secondary antibody to the rat IgG2a isotype,
if available. To minimize FcR-mediated binding of the secondary anti-Ig antibody, it is
preferable to use the F(ab )2 fragment rather than the whole immunoglobulin.

An alternative indirect staining method relies on the use of a biotin-conjugated primary


antibody, which is then detected with fluorochrome-conjugated streptavidin (SA). This
biotin-SA method is the preferred indirect staining approach, because reagent interactions,
which are often associated with the use of anti-Ig antibodies, are less of a concern. In
addition, there are commercially available SA conjugates to a variety of fluorochromes,
making SA useful in multicolor staining protocols. However, as with directly conjugated
MAbs, biotin-conjugated MAbs are not always available.

The use of secondary anti-Ig antibodies can be circumvented by labeling purified MAbs
with specific fluorochromes in the laboratory. Detailed methods for MAb conjuga-
tion are available on the Internet (Roederer, 1997), and conjugation kits can be ob-
tained from commercial vendors. The authors have had success using the Zenon Alexa
Fluor 488 Mouse IgG Labeling Kit (Molecular Probes, 2004a) to label unconjugated
MAbs. Using this antibody labeling kit, small quantities (≥0.4 µg) of primary MAbs
can easily be conjugated to fluorescent labels. Currently, the Zenon system can only
be used with mouse IgG1 , mouse IgG2a , mouse IgG2b , rabbit IgG, and human IgG
antibodies. Immunotoxicology

18.8.5
Current Protocols in Toxicology Supplement 23
Additional Materials (see also Basic Protocol 1)
Unconjugated primary MAb or biotin-labeled primary MAb
Fluorochrome-conjugated anti-Ig F(ab )2 fragment (for use with unconjugated
primary MAb) or fluorochrome-conjugated streptavidin (for use with
biotin-labeled primary MAb)
1. Stain cells with primary MAbs according to steps 1 through 9 of Basic Protocol 1.
If necessary, a combination of several fluorochrome-conjugated MAbs plus one non-
fluorochrome-conjugated MAb (preferably a biotinylated MAb) can be used in a single
MAb cocktail.

2. Resuspend cells in 50 µl PAB with gentle vortexing.


3. Add fluorochrome-labeled streptavidin or an appropriate secondary antibody. Vortex
lightly and incubate on ice for 20 min.
4. Add 150 to 200 µl PAB and centrifuge cells for 3 min at 200 to 1500 × g, 4◦ C.
Remove supernatant as in Basic Protocol 1, step 4.
5. Wash cells once in PAB as in Basic Protocol 1, step 9.
6. Resuspend cells in 500 µl PAB with gentle vortexing.
Alternatively, for samples prepared in microtiter plates, resuspend each cell pellet in
200 µl PAB. Next, using a multichannel pipettor, transfer the contents of each well to a
Titertube and add an additional 300 µl PAB, for a total volume of 500 µl. These Titertubes
can then be inserted into 12 × 75–mm plastic tubes for flow cytometric analysis.

7. Keep samples on ice until analyzed using a flow cytometer.


If stained cells cannot be analyzed on the day of preparation, they must be fixed for
longer-term storage (Support Protocol 2).

SUPPORT PREPARATION OF CELLS FROM LYMPHOID ORGANS


PROTOCOL 1
The processing of mouse lymphoid organs into single-cell suspensions is a fairly simple
procedure.

Materials
Lymphoid organ of interest
HBSS supplemented with 5% (v/v) FBS (5% HBSS; see recipe)
Endotoxin-screened distilled water for cell culture (Gibco)
10× HBSS (Sigma)
ACK lysis buffer (optional; see recipe)
60 × 15–mm untreated culture dish
25 × 75 × 1–mm frosted glass microscope slides
15-ml conical centrifuge tubes
12 × 75–mm disposable cell culture tubes
Coulter counter or hemacytometer
1. Transfer a freshly dissected lymphoid organ to a 60 × 15–mm culture dish containing
4 ml of 5% HBSS.
2. When ready for processing, place the lymphoid organ between the frosted ends of
two glass microscope slides. Disrupt the organ by gently pressing the frosted ends in
Immune Cell a circular motion until only the empty capsule remains.
Phenotyping
Using Flow
Cytometry

18.8.6
Supplement 23 Current Protocols in Toxicology
3. Transfer the resulting cell suspension to a 15-ml conical centrifuge tube. Rinse the
culture dish and capsule with a combined total of 3 ml of 5% HBSS and then add
this liquid to the cell suspension.
4. Centrifuge the cell suspension for 10 min at 200 × g, 4◦ C. Discard supernatant.

For spleen cell preparations


5a. Lyse RBCs by adding 4.5 ml cold distilled water (for cell culture) to the cell pellet
and resuspending with gentle vortexing for 10 sec. After vortexing, immediately add
0.5 ml of 10× HBSS to restore isotonicity, followed by an additional 5 ml of 5%
HBSS. Centrifuge cells as in step 4, discard supernatant, and resuspend the cell pellet
in 10 ml of 5% HBSS with gentle vortexing.
This procedure also lyses dead cells. If it is necessary to preserve dead cells (e.g., for
apoptosis studies), RBCs can be removed by (1) resuspending the cell pellet in 5 ml of a
standard ammonium chloride lysing solution (ACK lysing buffer; see recipe) and vortexing
briefly; (2) incubating the resuspended pellet for 5 min at room temperature; (3) adding
5 ml of 5% HBSS (inverting the tube several times to mix); and then (4) centrifuging the
cells for 10 min at 200 × g, 4◦ C, and discarding the supernatant.

In general, the viability of spleen cell suspensions will be substantially higher when the
hypotonic lysis method (>95%), as opposed to the ammonium chloride lysis method (85%),
is used.

6a. Let cell debris settle for 10 min on ice, and then remove the cell-rich supernatant and
transfer to a clean tube.

For thymus and lymph node cell preparations


5b. Resuspend the cell pellet in 10 ml of 5% HBSS with gentle vortexing.
6b. Let cell debris and clumps settle for 8 to 10 min on ice, and then remove the cell-rich
supernatant and transfer to a clean tube.
7. Determine cell concentration using a Coulter counter or hemacytometer (APPENDIX 3B).
Use 5% HBSS to adjust the final concentration of the cell suspension to 10–40 ×
106 cells/ml.

CELL FIXATION SUPPORT


PROTOCOL 2
If it is not possible to analyze freshly stained cells on the day of preparation, the cells must
be fixed in electron microscopy (EM)–grade formaldehyde (such as Ultrapure formalde-
hyde, Polysciences) prior to storage at 4◦ C. Fixation not only cross-links proteins to make
cells stable for storage but also inactivates the infectious activity of any viruses present.
Hence, cell fixation is highly advisable when handling biological specimens capable of
transmitting infection, and especially when handling human-derived cells.

Fixed samples should be analyzed within 5 days of fixation, since prolonged storage can
result in increased autofluorescence. This is usually not a problem when antigens that are
expressed at high levels are being analyzed. However, increased autofluorescence may
make it more difficult to resolve negative cells from dimly stained cells. Cell fixation
often leads to increased cell aggregation; therefore, it may be prudent to filter samples
through a 40-µm nylon mesh prior to running them on the flow cytometer.

Additional Materials (see also Basic Protocol 1)


1% formaldehyde: 10% (v/v) Ultrapure EM-grade formaldehyde (Polysciences)
diluted 1:10 in PAB (see recipe)
40-µm nylon mesh (Small Parts, Inc.; optional) Immunotoxicology

18.8.7
Current Protocols in Toxicology Supplement 23
1. Stain cells with MAbs according to steps 1 through 9 of Basic Protocol 1 or steps 1
through 5 of the Alternate Protocol.
2. Resuspend cell pellet in residual supernatant by gently vortexing.
Cells must be thoroughly resuspended prior to fixation. If gentle vortexing does not com-
pletely resuspend the cell pellets in the microtiter plate, use a multichannel pipettor
(equipped with plastic tips) to break up the pellets.

3. Add 200 µl 1% formaldehyde to each well and store cells on ice, protected from
light, for 30 min.
If necessary, cells can be stored overnight in 1% (v/v) formaldehyde at 4◦ C.

4. Prior to flow cytometric analysis, centrifuge cells 3 min at 1500 × g, 4◦ C. Remove


supernatant as in Basic Protocol 1, step 4, and resuspend the cell pellet in 500 µl PAB.
After resuspension in PAB, it may be desirable to filter samples through a nylon mesh to
remove cell aggregates, as this will prevent clogging of the flow cell nozzle.

BASIC STAINING OF PERIPHERAL BLOOD LEUKOCYTES


PROTOCOL 2
This protocol details the immunophenotyping of peripheral blood leukocytes, a process
often referred to as a “whole-blood” technique, as RBC lysis takes place after the sample
has been stained with specific MAbs. It is appropriate for the immunophenotyping of
blood cells from a number of animal species, including mouse, rat, horse, and dog.

Materials
Animals of interest
Sodium heparin, 250 IU per ml in PBS (APPENDIX 2A)
PBS supplemented with sodium azide and BSA (PAB; see recipe)
FcR-blocking Ig solution, 2 mg/ml (note that concentration differs from that used
in Basic Protocol 1)
Appropriate primary MAbs and (if necessary) secondary antibodies
FACS Lysing Solution (BD Biosciences), diluted 1:10 in distilled water according
to the manufacturer’s instructions
1-cc syringe equipped with 22-G needle
Coulter counter
Disposable 12 × 75–mm polystyrene tubes
NOTE: Staining conditions are similar to those noted in Basic Protocol 1. However, all
staining procedures should be performed in 12 × 75–mm polystyrene tubes, not microtiter
plates.

NOTE: Be sure to protect sample from light during incubation with fluorochrome-
conjugated MAbs.

Prepare peripheral blood leukocytes for staining


1. Using aseptic technique (APPENDIX 3B), collect blood (by cardiac puncture for mice)
in a 1-cc syringe containing sodium heparin.
A sufficient amount of heparin can be drawn into the syringe barrel by simply pulling the
plunger back to the 0.05-cc mark.
In order to reduce animal-to-animal variation, the same volume of blood should be col-
lected from each animal (for example, ∼500 µl from each mouse).
Immune Cell 2. Determine WBC concentration of blood sample using a Coulter counter.
Phenotyping
Using Flow
Cytometry 3. Add 50 to 100 µl blood to a 12 × 75–mm polystyrene tube.

18.8.8
Supplement 23 Current Protocols in Toxicology
4. Wash cells twice by adding 2 to 3 ml PAB to tube, centrifuging for 5 min at 1500 ×
g, 4◦ C, and removing supernatant.
This wash step minimizes artifactual staining of serum Igs with MAbs. Washing is per-
formed by adding 2 ml PAB and centrifuging cells for 5 min at 1500 × g, 4◦ C. This
centrifugation speed is required to produce a soft pellet of blood cells.
The supernatant is removed by carefully decanting it and then touching the lip of the tube
to an absorbent towel. Alternatively, the supernatant can be aspirated, taking care not to
disturb the soft cell pellet.

5. Resuspend blood cells in residual medium (∼100 µl) by gently vortexing.


Label with fluorochrome-conjugated MAbs
6. To block FcR-mediated binding of Igs, add 5 µl FcR-blocking Ig (10 µg) to the cell
suspension. Vortex cells lightly and incubate on ice for 10 min (Basic Protocol 1,
step 6).
7. Without removing the FcR-blocking reagent, add the appropriate MAbs (see
Support Protocol 4 for determination of optimal antibody concentrations) to the
cell suspension, vortex gently, and incubate on ice for 10 min.
All MAbs can be added concurrently in this step (Basic Protocol 1, step 7). It is highly ad-
visable to include a MAb to CD45 to help define the leukocyte gate during data acquisition
and analysis.

8. If all MAbs are fluorochrome-conjugated, proceed to step 11.


9. Add 2 ml PAB to sample and centrifuge for 5 min at 1500 × g, 4◦ C. Remove
supernatant and wash cells once with PAB as in step 4.
10. Add secondary antibodies to sample, vortex gently, and incubate on ice for 20 min.
11. Without removing or washing away the MAbs, add 2.0 ml FACS Lysing Solution,
1:10 dilution.
FACS Lysing Solution does not lyse nucleated erythrocytes, which are present in certain
animal species.

12. Vortex cells thoroughly and incubate for 10 min at room temperature.
13. Centrifuge sample for 5 min at 1500 × g, 4◦ C. Remove supernatant and wash cells
once with PAB as in step 4.
14. Resuspend cells in 500 µl PAB with gentle vortexing.
15. Keep samples on ice until analyzed using a flow cytometer.
The FACS Lysing Solution contains formaldehyde, so no further cell fixation is required
for longer-term storage.

ASSESSMENT OF CELL VIABILITY SUPPORT


PROTOCOL 3
Cell viability should routinely be assessed in all flow cytometric analyses. Because the
membrane integrity of dead cells has been compromised, most antibodies will freely
pass through such membranes and stain dead cells nonspecifically. This nonspecific
staining can make data difficult, if not impossible, to interpret and ultimately lead to
erroneous conclusions.

In general, cell viability is high (>95%) when lymphoid cells are taken directly from
animal tissues, unless the mechanical process used to create single-cell suspensions has
resulted in large numbers of dead cells. In contrast, the viability of cultured cells can be
Immunotoxicology
quite low, especially when these cells are chemically treated. In either case, to minimize
18.8.9
Current Protocols in Toxicology Supplement 23
cell death during sample processing, all cell preparations and staining procedures (in-
cluding wash steps) should be performed at 4◦ C or on ice and in the presence of BSA or
heat-inactivated FBS. The elimination of dead cells prior to staining is not recommended,
since any manipulation of the cell sample could lead to the inadvertent loss of viable cells.
Cells should be stained promptly after they are harvested and, if not fixed, analyzed in a
flow cytometer as soon as possible.

To assess cell viability, a sample of unstained cells is incubated with a DNA dye, such as
propidium iodide (PI) or 7-aminoactinomycin D (7-AAD); dead cells will stain positively
for either of these two nuclear dyes. Next, a data acquisition region is placed around the
positively stained cells. By color-eventing or backgating on the PI+ or 7-AAD+ cells
present, one can easily discern that most, but not all, dead cells exhibit lower forward
scatter (FS) and higher side scatter (SS) than do viable cells. A gating region is then
established around a cluster of viable cells (PI-negative) on the basis of their light scatter
profile. This gating region can be used for all subsequent samples, even if these samples
do not include a viability indicator. However, the best method for excluding dead cells
from data analysis is to use a vital DNA dye in all samples. Some of the more common
vital dyes used in multicolor analyses are PI, 7-AAD, TO-PRO-3 (Molecular Probes),
and pyronin Y(G) [PY(G)].
Materials
Nuclear staining compound dissolved in PBS (APPENDIX 2A): 200 µg/ml PI,
250 µg/ml 7-AAD, 250 µg /ml TO-PRO-3 (Molecular Probes), or 200 µg/ml
PY(G)
12 × 75–mm polystyrene tubes
Flow cytometer
1. Add 500 µl cells (1–2 × 106 ; unfixed and unstained) to a 12 × 75–mm polystyrene
tube.
2. Add 5 µl PI to tube.
Alternatively, add 4 µl 7-AAD, 4 µl TO-PRO-3, or 5 µl PY(G).
3. Incubate cells on ice for at least 5 min.
4. Analyze cells by flow cytometry.

SUPPORT TITRATION TO DETERMINE OPTIMUM ANTIBODY CONCENTRATION


PROTOCOL 4
The single most important factor in selecting an appropriate antibody is antibody quality.
Good antibodies are characterized by their high specificities and high binding affinities,
which allow specific staining to be distinguished from nonspecific staining. However,
even with high-quality antibodies, it is important to determine the optimal staining con-
centration (or titer) for each MAb. This titer will ensure that cells are stained under
saturating conditions. Under such conditions, specific fluorescence will not be readily
influenced by cell number or incubation time (Kantor and Roederer, 1997). The method
for titrating MAbs has previously been described by Stewart and Stewart (2001a).
Materials
Lymphoid cell suspension of interest (see Support Protocol 1)
Fc receptor (FcR)-blocking immunoglobulin (Ig) solution, 200 µg/ml
Fluorochrome-conjugated antibody solutions to be tested
Immune Cell Centrifuge fitted with a microtiter plate adapter
Phenotyping
Using Flow Vortex mixer fitted with a microtiter plate adapter
Cytometry Flow cytometer
18.8.10
Supplement 23 Current Protocols in Toxicology
1. Add 2 × 106 lymphoid cells to each of five wells in a 96-well microtiter plate.
2. Centrifuge cells for 3 min at 200 to 1500 × g, 4◦ C, in a centrifuge fitted with a
microtiter plate adapter.
3. Block FcR-mediated binding by adding 50 µl FcR-blocking Ig to each well, vortexing
gently using a vortex mixer fitted with a microtiter plate adapter, and then incubating
samples for 10 min on ice.
4. Without removing the FcR-blocking Ig added in step 3, add 0.125, 0.25, 0.5, 1.0,
and 2.0 µg of MAb, respectively, to the five wells containing lymphoid cells. Vortex
cells gently and incubate for 10 min on ice.
5. Acquire immunofluorescence data using a flow cytometer
6. Create a two-parameter histogram of forward scatter versus side scatter and establish
an appropriate gating region that encompasses the majority of viable cells.
7. Display fluorescence data from viable cells in single-parameter histograms. Establish
one region that encompasses the negative population and a second region that brackets
the positive population. Determine the median channel of fluorescence (MCF) for
each region.
8. Compute the signal-to-noise (S/N) ratio, treating the positive population as the signal
(S) and the negative population as the noise (N). Plot the calculated S/N ratios as a
function of MAb concentration.
9. Identify the optimal MAb titer (i.e., the titer that produces the highest S/N ratio).
Once the optimal titer is determined, it is not necessary to increase the amount of antibody
used for staining unless the number of cells per sample exceeds 20 million. Even then,
the amount of antibody needed for staining will only increase by a factor of two or
three. However, it cannot be stressed enough that time and effort should be taken to
optimize staining conditions (staining temperature, antibody concentration, staining time,
and cell number) prior to any study. Doing so can also save money, as the manufacturer’s
recommended staining concentration is often higher than necessary.

REAGENTS AND SOLUTIONS


Use Milli-Q-purified water or equivalent for all recipes and protocol steps. For common stock
solutions, see APPENDIX 2A; for suppliers, see SUPPLIERS APPENDIX.

ACK lysing buffer


0.15 M NH4 Cl
10 mM KHCO3
0.1 mM disodium EDTA
Adjust pH to between 7.2 and 7.4 using 1 M HCl
Filter sterilize using a 0.2-µm filter
Store at room temperature
If the buffer is stored for an extended period, check its pH before use.

HBSS, pH 7.2, supplemented with 5% (v/v) FBS


1× Hanks’ balanced salt solution (HBSS; Sigma) containing:
1 mM N-2-hydroxyethylpiperazine-N -2-ethanesulfonic acid (HEPES)
5% (v/v) fetal bovine serum (FBS), low-endotoxin, characterized (HyClone)
1 mM sodium pyruvate
Adjust pH to 7.2 using 1 N NaOH
Immunotoxicology
Store up to 1 month at 4◦ C
18.8.11
Current Protocols in Toxicology Supplement 23
PBS, pH 7.2, supplemented with sodium azide and BSA
Dulbecco’s PBS, calcium- and magnesium-free (e.g., Life Technologies),
containing:
0.1% (w/v) sodium azide
1% (w/v) BSA
Adjust pH to 7.2 using 1 N NaOH or 1 N HCl
Filter solution through 0.22-µm filter paper
Store up to 1 month at 4◦ C
EDTA (1 mM) can be added to reduce cellular aggregation.
CAUTION: Be extremely careful when handling sodium azide.

COMMENTARY
Background Information MAb, one could select a secondary MAb spe-
cific for rat IgG1 , if available.
Secondary anti-Ig antibodies In all cases, it is necessary to determine the
Although indirect staining with secondary
optimum antibody titer as well as assess the
fluorochrome-conjugated anti-Ig antibodies
binding specificity of the secondary anti-Ig an-
is the least desirable staining method, due
tibody prior to a study. First, the secondary
to problems with nonspecific staining, there
antibody should not react with any cellular
are certain precautions that can be exercised
epitope. This can be assessed by simply incu-
to minimize these problems. The secondary
bating the cells of interest with the secondary
anti-Ig antibody is usually a polyclonal anti-Ig
antibody alone. The signal produced by this
antibody that is specific for a large number of
staining should be nearly identical to the signal
epitopes on the primary antibody. Since poly-
yielded by unstained cells. If not, the reagent
clonal secondary antibodies will be directed
is inappropriate and should be replaced.
against all the Ig isotypes (e.g., IgG, IgA,
Second, if this secondary antibody is used in
IgM) found in the serum of the animal from
multicolor analyses, it should not cross-react
which they were derived, these antibodies
with any other MAb used for staining. To
should be purified by affinity chromatography
test for potential reagent interactions, incubate
so that their binding is restricted to the heavy
two samples of cells with the fluorochrome-
chain of the appropriate Ig isotype. The
conjugated MAb, followed by washing. Next,
binding of the secondary anti-Ig antibody can
add the secondary anti-Ig antibody to one sam-
be further restricted by selecting a reagent
ple and PAB to the other sample. The fluo-
with no light-chain activity (i.e., one that is Fc
rescence pattern of the sample stained with
fragment–specific). This will further ensure
the secondary anti-Ig reagent should be nearly
that the anti-Ig antibody is highly specific for a
identical to the pattern produced by the sample
particular class of Ig (e.g., IgG) and thus min-
stained with PAB. If the fluorescence pattern
imize cross-reactions with other Ig isotypes.
is different, the secondary antibody in question
In addition, it is essential to use the F(ab) or
should not be used in multicolor analyses.
F(ab )2 fragment of the affinity-purified sec-
ondary antibody to minimize FcR-mediated Fluorochrome choice
binding and, hence, reduce background stain- Fluorochrome choice is highly contingent
ing. For example, a goat F(ab )2 anti–mouse on the availability of the laser lines avail-
IgG antibody (heavy and light chain–specific, able on a particular flow cytometer. For in-
purified by affinity chromatography) is stance, the Beckman Coulter XL flow cytome-
specific for the heavy chain of mouse IgG but ter is equipped with a single argon ion laser
will also bind to other Igs that have the same tuned to a wavelength of 488 nm, the most
light chain as IgG (e.g., IgA, IgM). Therefore, common wavelength used in flow cytometry.
this reagent is not specific for mouse IgG at This instrument can handle up to four fluo-
all. The authors have had great success using rescent emissions, all from dyes that can be
polyclonal secondary antibodies purchased excited by the 488-nm wavelength, but it can-
from Jackson ImmunoResearch Laboratories. not process the signal generated by a MAb
The binding of a secondary antibody can be conjugated to allophycocyanin (APC), which
Immune Cell
Phenotyping further restricted to a specific Ig isotype and must be excited by a 633-nm laser line. How-
Using Flow subclass. For example, if a rat IgG1 MAb spe- ever, APC signals can be processed on any
Cytometry cific for CD4 were being used as the primary benchtop analyzer, such as the FACScalibur
18.8.12
Supplement 23 Current Protocols in Toxicology
(BD Biosciences) or the FC500 (Beckman particularly sensitive to photodegradation.
Coulter), that is equipped with a HeNe laser. This phenomenon can easily be recognized by
For multicolor analyses, there are a number an increase in the PE signal and a concomitant
of factors to be considered when selecting a decrease in the Cy5 or Cy7 signal. Use of
MAb conjugated to a particular fluorochrome. cyanine tandem dyes can result in increased
This selection is particularly important when background staining, as these dyes can bind to
using antigens that are expressed at low monocytes and B cells. However, this binding
density, as some fluorochromes have higher is highly species-dependent; for instance,
absorption coefficients and/or higher quantum background binding can be high in mice
yields than others, making them more suitable with autoimmune disorders. Thus, again, it is
for detecting weakly expressed antigens. In prudent to assess the background staining of
order to achieve the highest possible staining the reagents used prior to a large study.
intensity, select antibodies conjugated to the
brightest possible fluorochromes, e.g., in Multicolor staining combinations for
order, from brightest to dullest—PE, PE-Cy5, routine immunophenotyping of murine cells
PE–Texas Red, APC, and FITC. In general, Table 18.8.1 presents examples of possible
the fluorescence emitted by MAbs conjugated staining strategies that can be used to pheno-
to PE, PE-Cy5, and APC is 5 to 10 times type various cell populations in the mouse. It
brighter than the fluorescence emitted by is becoming increasingly clear that no single
FITC conjugates. PE is often the fluorochrome marker can be used to specifically identify a
of choice when examining the expression of unique subset of cells. For instance, expression
an antigen that is expressed at low levels (e.g., of CD4 and CD8a is not restricted to T cells, as
CD25, CD69) or detecting the presence of these molecules are also expressed on subsets
intracellular cytokines. of dendritic cells (CD11c+ ). In addition, CD4
New fluorochromes are continually being is expressed on a subset of natural killer (NK)
developed, and they can be used in addition cells. Thus, in order to ensure that only T
to or instead of the dyes mentioned above. cells are analyzed, a MAb specific for CD3e,
Molecular Probes (2004b) has produced a new a marker that is expressed on thymocytes
series of dyes, called the Alexa Fluor dyes, and mature T cells, can be included in the
that exhibit more intense fluorescence than MAb cocktail. The authors have also found
do other spectrally similar conjugates. For that the correlated expression of two markers,
example, Alexa488, whose fluorescence spec- CD11b and Gr-1, is needed to phenotype either
trum is nearly identical to that of fluorescein macrophages or granulocytes in mouse spleen.
isothiocyanate (FITC), is considered by many Both of these cell types express CD11b and
to be the best FITC-like reagent. A number Gr-1; however, the level of Gr-1 is lower on
of MAbs conjugated to Alexa488 can be macrophages than on granulocytes. The two
obtained from commercial sources. If the ex- populations are more clearly resolved by in-
periment necessitates the use of a second-step cluding additional markers that are expressed
reagent, the authors use fluorochrome-labeled on the macrophage, such as F4/80, CD86, or
SA, which is readily available from commer- CD54 (Choi et al., 2003). It is essential to
cial sources, to stain biotin-labeled MAbs. recognize that many markers can be expressed
For simple one- or two-color analyses, the at varying levels on different cell types.
second-step reagent of choice for the authors
is PE-labeled SA, which will stain at the
highest possible level. For multicolor studies,
Critical Parameters
the authors routinely use a tandem conjugate Autofluorescence
PE–Texas Red–labeled SA. It is possible that Cellular autofluorescence is most often
PE-Alexa610-labeled SA, by virtue of its low associated with myeloid cells, due to the
background staining, may be a better option presence of intracellular flavins in such cells.
than the PE–Texas Red fluorochrome, which These flavins are easily excited by the 488-nm
can exhibit the high background staining that laser line, with peak emission occurring
is typical of Texas Red dyes. The authors around 525 nm. Therefore, any signal gener-
rarely use FITC-labeled SA but have had great ated by autofluorescent cells will be processed
success with Alexa488-labeled SA, which is by the same photomultiplier tube (PMT) that
far superior to its FITC-labeled counterpart. processes FITC fluorescence. It is obvious
All MAbs should be stored in the dark, that the presence of highly autofluorescent
as most fluorochromes are photosensitive. cells can contribute an unwanted addition
Immunotoxicology
PE-Cy5 and PE-Cy7 tandem conjugates are to the FITC signal. Autofluorescence can
18.8.13
Current Protocols in Toxicology Supplement 23
Table 18.8.1 Examples of Possible Antibody Combinations for Staining Mouse Leukocytes
Using Five Fluorochrome-Labeled Monoclonal Antibodies in a Staining Cocktail, with Analysis
Performed on an FC500 Flow Cytometer (Beckman Coulter) with Dual Laser Excitation at 488
nm and 633 nma

Cell type(s) Possible combination of stains

T cells, NK cells FITC-anti-CD4


PE-anti-CD3e
Biotin-anti-NK1.1b
PE-Cy5–anti-CD8c
PE-Cy7–anti-CD45d,e
Viable T cells FITC-anti-CD4
PE-anti-CD3e
Biotin-anti-NK1.1b
7-AADf
PE-Cy7–anti-CD8
Granulocytes, dendritic cells, B cells, macrophages FITC-anti-Gr-1
PE-anti-CD11c
ECD-anti-B220g
PE-Cy5–anti-CD11b
PE-Cy7–anti-CD45
Activated T cells FITC-anti-CD8
PE-anti-CD25
ECD-anti-CD4
PE-Cy5–anti-CD62Lh
PE-Cy7–anti-CD8
a Abbreviations: 7-AAD, 7-aminoactinomycin D; APC, allophycocyanin; FITC, fluorescein isothiocyanate; MAb, mon-
oclonal antibody; NK, natural killer; PE, phycoerythrin.
b This biotinylated MAb necessitates the use of streptavidin labeled with PE–Texas Red or equivalent.
c PE-Cy5-labeled MAb could be replaced by a MAb conjugated to APC or equivalent.
d PE-Cy7-labeled MAb could be replaced by a MAb conjugated to APC-Cy7 or equivalent.
e CD45 is a pan-leukocyte marker that is very useful in discriminating leukocytes from contaminating red blood cells,
especially in the phenotyping of peripheral blood cells. A gating region can be established on the CD45+ leukocyte
population as defined in a two-parameter histogram of side scatter versus CD45 expression.
f 7-AAD is a DNA dye used as a viability indicator in unfixed cell samples. Its peak emission wavelength is 660 nm.
g ECD is the trade name for the PE–Texas Red tandem conjugate produced by Beckman Coulter. Very few mouse
marker–specific MAbs that are directly conjugated to this fluorochrome are available.
h Because CD62L can easily be shed from the surface of cells, it is imperative to keep cells on ice throughout staining
and data acquisition. The authors recommend that samples stained with anti-CD62L be kept on ice until loaded manually
onto the flow cytometer for data collection. The use of a multisample carousel is not recommended when analyzing cells
stained for CD62L, since cells cannot be kept cold while on such a device.

be particularly bothersome when measuring MAbs that are conjugated to APC or APC-
weak FITC fluorescence signals. If there are Cy7, as these dyes can be excited by a HeNe
excessive numbers of myeloid cells in the laser and emit at wavelengths greater than
experimental samples, the resulting high level 600 nm, beyond the emission spectrum for cel-
of autofluorescence may make it difficult to lular autofluorescence. When immunopheno-
interpret data. A sample of unstained cells typing lymphocytes in a sample containing el-
can be used to determine the contribution of evated numbers of myeloid cells, one could
cellular autofluorescence to the background include a MAb that specifically identifies the
fluorescence signal as well as the minimum myeloid cells (such as one specific for Gr-1, a
level of fluorescence exhibited by cells. marker of myeloid/granulocyte lineage). The
There are several approaches that can be use of such an antibody allows one to gate
Immune Cell
Phenotyping used to circumvent the problems associated out granulocytes and macrophages, thereby
Using Flow with cellular autofluorescence. When study- eliminating cellular autofluorescence from
Cytometry ing myeloid cells, it may be prudent to use analysis.
18.8.14
Supplement 23 Current Protocols in Toxicology
Blocking FcR-mediated binding The fluorescence pattern of the isotype control
There are two ways that an antibody can should be nearly identical to that of the
specifically bind to cells. Antibodies can bind autofluorescence control. When this is not the
to a specific antigen (epitope-specific binding) case, it is indicative of an improperly titrated
through their antigen-binding sites, the F(ab ) MAb, a poor-quality MAb, or both. This
fragment, or they can bind to myeloid cells could have a profound effect on one’s ability
that express receptors for the Fc portion of the to interpret positive fluorescence, especially
Ig. Even though FcR-mediated binding is not when studying antigens that are expressed
antigen-specific, it is highly specific and needs either transiently or at very low levels.
to be eliminated in order to assess antigen- In general, the authors have found that some
specific staining. isotype Igs (e.g., IgM, IgG2b ) exhibit more
FcR should be blocked with normal Ig prior background staining than others, which can
to staining with epitope-specific MAbs. The complicate the analysis of antigens expressed
authors typically incubate cells for 10 min at very low levels. This is particularly rele-
at 4◦ C with purified normal Ig (Jackson vant in analysis of intracellular antigens. In
ImmunoResearch Laboratories) taken from these cases, a cold block control is performed
the same species that produced the antibodies to verify antibody specificity. This can eas-
used for staining. For instance, if a rat IgG2a ily be accomplished by comparing the fluo-
anti-CD4 MAb is used, FcR-mediated binding rescence of two cell samples, with one be-
should be blocked with an excess of purified ing pretreated with a cold blocking MAb (i.e.,
rat IgG. The use of 10 µg of normal IgG in a the same clone as the experimental MAb, but
staining volume of 50 µl (final concentration, with no conjugate) for 15 min prior to the ad-
200 µg/ml) should be sufficient to block FcR. dition of the fluorochrome-conjugated MAb.
After 10 min of blocking, the MAbs are added The binding of the fluorochrome-conjugated
directly to the samples without removing the MAb should be completely inhibited by the
FcR block, so that epitope-specific staining can cold block. For example, to assess the expres-
proceed in the presence of FcR-block. Another sion of CD25, which can be present at low
approach to blocking FcR is to incubate cells levels on activated T cells, cells are pretreated
with an unconjugated MAb that is specific for with unlabeled purified anti-CD25 MAb (at
the FcR (CD16/CD32). It should be noted that a 3-fold higher concentration compared with
this approach is more expensive than the for- the conjugated MAb) for 15 min prior to the
mer approach. In any case, do not block FcR if addition of the fluorochrome-conjugated anti-
there is interest in assessing FcR expression. CD25 MAb. This sample should represent a
true negative control for CD25 staining. Back-
Isotype controls ground staining for cell surface expression of
Isotype controls are also used to determine CD69, another T cell marker that is transiently
the minimum level of cellular fluorescence up-regulated upon activation, can be assessed
following staining and to assess the effective- similarly. Due to the complexities involved in
ness of FcR blocking. An isotype control must multicolor analyses, it is prudent to invest a lit-
match the primary MAb in terms of isotype tle time to test these conditions prior to a large
and subclass, as well as fluorochrome label and study.
concentration. For example, the appropriate Background staining exhibited by cells
isotype control for cells incubated with 0.5 µg stained with multiple reagents is very different
FITC-labeled rat IgG2a MAb (specific for from that seen in unstained cells or isotype con-
mouse CD4) is 0.5 µg FITC-labeled normal trols, due to the broadening artifacts produced
rat IgG2a . The appropriate isotype control by compensation (see below) and/or reagent-
for cells stained with biotin-labeled rat IgG1 reagent interactions. Therefore, the best neg-
MAb (specific for CD8) is treatment with ative control for any given marker in a mul-
biotin-labeled normal rat IgG1 , followed by ticolor stain is a sample of cells stained with
incubation with fluorochrome-conjugated SA. all but one reagent (commonly referred to as
For multicolor analyses, one isotype “fluorescence minus one,” or FMO). For ex-
control sample can be set up to assess all ample, when assessing CD25 expression on
the MAb isotypes in a single tube. It is not antigen-specific D011.10 T cells, the best neg-
necessary to analyze each isotype control ative control for CD25 expression is a cocktail
separately. For example, a sample of cells can containing MAbs to CD3, CD4, and KJ-126
be incubated, in the presence of an FcR block, (to identify the antigen-specific T cells in the
with a cocktail of isotype-matched Igs for each D011.10 model) plus an isotype control for the
Immunotoxicology
specific MAb used in the staining procedure. CD25 MAb. In this way, the level of negative
18.8.15
Current Protocols in Toxicology Supplement 23
staining for CD25 expression can be assessed from the perspective of designing and analyz-
by gating on the antigen-specific T cells. ing multicolor experiments. Investigators need
to be able to recognize data that has resulted
Dead cells
from improper compensation, as failure to do
Cell viability must be assessed in each
so can easily result in data misinterpretation.
experimental study. Since most MAbs freely
Any spillover signal not completely compen-
pass through the cell membranes of dead and
sated (i.e., undercompensated) out of a PMT
damaged cells, dead cells will stain positively
can result in an overestimation of the pos-
even if there is no specificity for a particular
itive population. Proper compensation is al-
MAb. This nonspecific fluorescence can inter-
ways essential when measuring antigen den-
fere with data analysis, and if ignored, it can
sity by MCF.
result in erroneous interpretation of the data.
Compensation is often considered a process
For exclusion of dead cells, vital DNA dyes
by which unwanted fluorescent signals are
such as PI, 7-AAD, and TO-PRO-3 can be
“subtracted” from the true fluorescent signal.
added to unfixed cell samples prior to analysis.
However, compensation is not a subtraction
These dyes cannot be used to assess the viabil-
process, but rather a straightforward applica-
ity of cells that are permeabilized (as for intra-
tion of linear algebra by which a proportion of
cellular staining) or fixed, since they will leak
the unwanted signal is eliminated from the true
out of fixed cells. On the other hand, the vital
fluorescent signal. For some fluorochrome
dye ethidium monoazide (EMA; final concen-
combinations, such as FITC and PE-Cy5 or
tration, 1 to 5 µg/ml) can be used to label dead
APC, there is little, if any, overlap in spectral
cells prior to fixation (Reidy et al., 1991). Once
emissions; therefore, compensation is negligi-
inside a dead cell, EMA can be photochemi-
ble. However, there is considerable overlap in
cally cross-linked to the DNA by exposure to
the emission spectra of the PE, PE–Texas Red,
visible light. This cross-linking allows EMA
and PE-Cy5 dyes, enough to make compen-
to be retained by fixed cells.
sation quite challenging even for investigators
Compensation who routinely perform multicolor analyses.
One of the most critical issues in perform- In order to set up compensation properly, a
ing multicolor phenotyping is dealing with sample of cells must be stained with a repre-
overlaps in the spectral emissions of different sentative reagent for each fluorochrome used
fluorochromes. Once excited by the laser, a in the experiment. The number of compensa-
fluorochrome will emit a broad band of light. tion controls will be equivalent to the number
Even though a band-pass filter is often placed of fluorochromes used in staining. For exam-
in front of a PMT to restrict the wavelengths ple, five-color staining requires five compen-
of light that are transmitted to the tube, it is sation controls, one for each fluorochrome. It
nearly impossible to exclude all of the light is not necessary to use the same MAbs in the
emitted by the other fluorochromes used in experiment as in the compensation controls.
multicolor staining. Because of these spectral This is especially true when a particular exper-
overlaps, each fluorochrome will contribute imental reagent is expected to stain just a small
an unwanted light signal to several PMTs that subset of cells. For example, the frequency of
are not assigned to detect that fluorochrome. antigen-specific T cells (KJ1-26+ CD4+ ) adop-
For example, there is significant overlap in tively transferred in the D011.10 model can be
the spectral emissions of FITC and PE, so as low as 0.3% one day after injection with
much so that some of the FITC emission ovalbumin. Thus, it would be very difficult, if
spills over into the PE PMT and it must be not impossible, to properly set compensation
eliminated from this PMT before analyzing levels using the conjugated MAb that stains
PE fluorescence. By the same accord, PE KJ1-26+ cells. What is important is using the
contributes an unwanted signal to the FITC brightest possible reagents for compensation
PMT, albeit at much lower levels, and this controls. Therefore, the authors routinely set
signal must be eliminated from the FITC PMT. up compensation controls using CD45, CD8a,
If not eliminated, these spillovers will make a CD19, or CD45R MAbs conjugated to the ap-
“false” contribution to the data. The process by propriate fluorochromes. These markers are
which these false contributions are eliminated expressed at high levels on most immune cells,
electronically is called compensation. making it much easier to properly set com-
Immune Cell Compensation is one of the least understood pensation levels. In order to set up compensa-
Phenotyping processes in multicolor analyses. It is impor- tion properly, the compensation control sam-
Using Flow tant to consider compensation not only from ple must contain at least two populations—
Cytometry
the standpoint of applying it correctly but also specifically, one bright population and one
18.8.16
Supplement 23 Current Protocols in Toxicology
negative or not-so-bright population. When ex- example, the Beckman Coulter FC500 and XL
amining spleen cells, there will be two distinct flow cytometers are capable of handling the
populations that stain for MAbs to CD8a and pairwise compensation involving FITC and
CD45R; one is positive, and the other is nega- PE-Cy5, but the BD FACSCalibur is not. It can
tive. Because CD45 stains all leukocytes, it is be argued that, in general, there is minimal, if
necessary to add an aliquot of unstained cells any, FITC emission that spills over into the PE-
to the CD45 compensation control after the Cy5 PMT. However, when using carboxyflu-
staining process but just prior to flow analysis orescein diacetate succinimidyl ester (CFSE),
to artificially create a negative population. a low but measurable signal from this dye can
There are two ways to perform compen- spill over into the PE-Cy5 PMT. Thus, when
sation. Hardware compensation is performed using the BD FACSCalibur, in lieu of hardware
during data acquisition, whereas software compensation, one must rely on software com-
compensation is performed during data analy- pensation to eliminate unwanted CFSE signal
sis. Because of the complexity of five-color from the PE-Cy5 detector.
analyses, the authors routinely collect flow It is possible to minimize the need for com-
cytometric data without performing hardware pensation by carefully selecting reagents that
compensation in such analyses. Instead, prior are conjugated to fluorochromes whose emis-
to analyzing the experimental data using of- sions have little or no spectral overlap. For
fline software, the data collected from compen- two-color analyses, the authors often select
sation controls are used to perform software MAbs that are conjugated to FITC and ei-
compensation using WinList software (Ver- ther APC or PE-Cy5, since there is little, if
ity). The authors have found no disparity in the any, overlap between the emission spectrum
results obtained using software compensation of FITC and the emission spectra of the lat-
as compared with hardware compensation. ter two dyes. Beyond two-color analyses, flu-
Compensation controls must be evaluated orochrome choice gets more complicated with
in every possible pairwise combination us- each additional fluorochrome. In general, PE–
ing two-parameter histograms. For example, Texas Red–conjugated MAbs are used by the
in studies using FITC, PE, and PE-Cy5 fluo- authors only when performing five-color im-
rochromes in a single sample, the FITC com- munophenotyping, as this fluorochrome has
pensation control must be evaluated using considerable spectral overlap with both PE
the following two-parameter histograms: FITC and PE-Cy5 emissions, making compensation
versus PE and FITC versus PE-Cy5. Two- quite challenging. Of lesser importance, there
color samples require the assessment of just are very few MAbs that are directly conju-
one pairwise combination, three-color samples gated to PE–Texas Red, and two-step staining
require three combinations, four-color samples procedures are therefore often required. If a
require six combinations; and five-color sam- HeNe laser is available, the authors often se-
ples require ten combinations. Thus, it should lect APC- or APC-Cy7-labeled reagents over
be readily apparent that the complexity of com- PE-Cy5 conjugates.
pensation increases substantially with the ad-
dition of each fluorochrome. To properly set Control samples
compensation levels, the center of the positive In order to ensure that data are collected and
cell population (i.e., as determined by the me- interpreted correctly, several control samples
dian fluorescence intensity) is lined up with the must be faithfully evaluated prior to running
center of the negative cell population. This is the experimental samples. These include con-
accomplished by adjusting compensation until trols to assess cellular autofluorescence, non-
the median channel for the positive population specific binding, FcR-mediated binding, and
is equal to the median channel for the negative compensation.
population. It is important to ensure that the Sometimes, the number of essential control
fluorescence distribution for the negative pop- samples exceeds the number of experimental
ulation is not piled up on the baseline axis, as samples. However, it is only through the proper
this will make it nearly impossible to determine use of all appropriate control samples that data
the median channel and, thus, very difficult to can be collected and interpreted correctly. Con-
set up compensation properly. An in-depth dis- trols can be performed for each animal, but
cussion on compensation can be found online this can easily lead to excessive numbers of
(Roederer, 2000). control samples. For large studies, a pool of
Not all flow cytometers can handle all cells is created for each treatment group, and
the possible pairwise combinations that may this pool is used to test for autofluorescence
Immunotoxicology
be necessary for hardware compensation. For and isotype staining. In this way, a pool will
18.8.17
Current Protocols in Toxicology Supplement 23
contain representative cells from each animal introduced by the instrument can easily be
in a given treatment group. For example, in minimized. The instrument’s overall perfor-
an experimental study in which the effects of mance should be routinely evaluated prior
three different dose levels of a chemical are ex- to data acquisition using standardized beads,
amined, there will be four pools of cells, each such as Beckman Coulter Flow-Check beads.
representing a particular treatment group (one These beads will assess the instrument’s flu-
for the vehicle control group and one for each idics and optics to ensure proper alignment
of the three dose levels). Each treatment pool of the laser with the sample stream. Once
will have its own autofluorescence control and the fluidics and optics have been checked,
its own isotype control, resulting in a total of the authors routinely run a sample of Flow-
four autofluorescence controls and four iso- Set beads (Beckman Coulter) to further stan-
type controls. dardize the instrument’s settings. These beads
To assess autofluorescence, a sample of un- mimic the immunofluorescence exhibited by
stained cells (with no MAbs added) is pro- human and mouse cells and can be used at the
cessed in a similar fashion as for all exper- voltages required for immunophenotypic anal-
imental samples. This sample should always yses. The voltages and gain settings for each
be analyzed first in an experiment. To begin, photodetector are adjusted to position the flu-
the voltages for the photodetectors collecting orescence emitted by the beads within a pre-
the light-scatter signals (both forward scatter scribed region, a process commonly referred to
and side scatter) are adjusted to define the cells as channel positioning. In this way, any appar-
of interest. Second, the voltage of each PMT is ent change in marker expression over time can
adjusted to place the negative cell population be expected to be biologically relevant, and not
in the first log decade of the histogram. It is im- a result of changes in the instrument.
portant to apply enough voltage to ensure that Once it has been established that the instru-
the majority of cells are off of the baseline; this ment is performing under the expected exper-
assures that any positive staining will fall be- imental conditions, negative controls (autoflu-
yond the first log decade. In essence, this con- orescence and isotype) can be analyzed to en-
trol sample provides the baseline representing sure that the negative population falls within
the minimum (or negative) fluorescence exhib- the first log decade for each channel being mea-
ited by cells. sured. Next, compensation controls should be
The next control to be analyzed is the iso- performed to remove any unwanted fluores-
type control. The fluorescence pattern of the cence from the various PMTs due to overlaps
isotype control should be similar to that of in the spectral emissions of the fluorochromes
the unstained control if FcR-mediated binding used. It should be noted that improper com-
has been blocked effectively. If not, one would pensation can result in false-positive or false-
need to evaluate the isotype controls used in the negative results, so great care should be exer-
experiment. Like the autofluorescence control, cised when performing these controls.
the isotype control also determines the mini- Once the instrument’s settings have been
mum level of fluorescence that is exhibited by established, the authors routinely run a con-
cells. trol sample, referred to as the verification con-
For multicolor experiments, compensation trol, in order to determine whether the sys-
controls must be analyzed in order to account tem will provide accurate information. This
for any spectral overlap not removed by filters. control sample is stained with a set of MAbs
The number of compensation controls will be that are conjugated to the same fluorochromes
equivalent to the number of fluorochromes used in the experimental study but are specific
used in staining. Compensation is greatly sim- for well-recognized markers. For example, in
plified in four- or five-color analyses when the a five-color staining experiment (using MAbs
PMTs are balanced, with their voltages set at conjugated to FITC, PE, PE–Texas Red, APC,
nearly identical levels. However, it is crucial to and APC-Cy7), the verification tube would
remember that once compensation is set, any contain MAbs specific for CD4, CD8, CD3,
change in the PMT voltages will necessitate CD45R, and Gr-1, with each of these MAbs
repetition of the entire compensation process. being conjugated to one of the fluorochromes
used in the study. In general, spleen cells from
Standardization of flow cytometric data a naive mouse are used in these controls. Not
Immune Cell There are three sources of error that can be only are the authors familiar with the stain-
Phenotyping introduced during immunophenotypic analy- ing profiles of these markers, but they have
Using Flow sis: specifically, instrumental, technical, and established a historical record that they can
Cytometry
biological. However, the amount of error use for comparison with the observed relative
18.8.18
Supplement 23 Current Protocols in Toxicology
frequencies. In short, this verification control is splenic suspensions often exceeds 97%. When
expected to yield easily recognizable data that immunophenotyping blood cells, the authors
enable the authors to make any necessary fi- routinely include a MAb to the pan-leukocyte
nal changes to compensation values. Once the marker CD45, which allows them to set a
verification control has produced the expected gate around all the leukocytes present, thereby
results, one can be confident that the flow cy- eliminating, or gating out, residual RBCs and
tometer will provide accurate results for the platelets; this is most often performed by set-
experimental samples. ting a gate around the appropriate cell clusters
as defined by their CD45-versus-SS profiles.
Data analysis Two days after adoptive transfer into F1
The authors routinely acquire listmode data mice, one can easily identify a small but dis-
on 10,000 cells of interest when performing tinct cluster of cells that represent the donor
multiparametric analysis. For example, when CD4+ cells (Fig. 18.8.1B). These cells are
immunophenotyping spleen cells for lineage- identified by their expression of CD4 and their
specific markers (such as markers of B cell, lack of expression for H2Dd . As shown in this
T cell, macrophage, or granulocyte lineage), two-parameter histogram, the majority of cells,
they typically collect flow cytometric data on which are the host cells, express H2Dd . Only
10,000 viable spleen cells. If the authors are a small population of cells lacks H2Dd expres-
interested in analyzing cell surface markers sion, representing the donor T cells. An ellip-
gated on a discrete subset, such as antigen- tical region has been set to establish a gate for
specific CD4+ cells in the D011.1 model these CD4+ donor cells so that their expres-
(which can represent as little as 0.5% to 4% sion of the T cell activation markers CD28,
of the total population), they collect data on CD62L, and CD25 can be monitored through-
10,000 antigen-specific CD4+ cells. Following out the course of the GvH response.
data acquisition, flow cytometric data can be Gating on the donor CD4+ cells (10,000)
displayed in either single-parameter or two- as shown in Figure 18.8.1B, one can gener-
parameter histograms to monitor the cells of ate single-parameter histograms that illustrate
interest. In this way, gating and analysis strate- the expression of the T cell activation markers
gies can be fine-tuned. (Fig. 18.8.1C). The immunofluorescent profile
Figure 18.8.1 illustrates one approach to the for each marker is illustrated in the filled his-
analysis of five-color flow cytometric data. The tograms, while the open histograms illustrate
data represented in this figure are derived from negative staining as determined by their appro-
studies monitoring the events of T cell activa- priate isotype controls. Two populations can
tion in the graft-versus-host (GvH) response. easily be distinguished on the basis of the dif-
T cells (both CD4+ and CD8+ ) were purified ferential expression of CD62L, CD62Lpos , and
from the spleens of C57Bl/6 mice (H-2b ; re- CD62Lneg . However, the staining profiles for
ferred to as donors) and injected into C57Bl/6 CD25 or CD28, as seen in single-parameter
(H-2b ) × DBA/2 F1 mice (H-2d ; referred to histograms (Fig. 18.8.1C), are more homoge-
as hosts). The donor T cells recognize the neous.
H-2d antigens contributed by the DBA/2 strain Figure 18.8.1D illustrates the power of mul-
in the F1 host and become activated. Two days tiparametric analysis. Multiple subpopulations
after the injection of these donor T cells, the can be resolved by displaying the correlated
F1 mice were sacrificed, and their spleens were expression of two markers in two-parameter
processed into single-cell suspensions. Spleen histograms. While there is a hint of two pop-
cells were then stained simultaneously with a ulations in each of the single-parameter his-
cocktail of five MAbs specific for CD4, H-2Dd , tograms for CD25 and CD28 expression, one
CD28, CD25, and CD62L. could easily identify two cell clusters by cor-
As shown in Figure 18.8.1A, a cluster of relating the expression of these two markers
spleen cells was first identified by its light scat- in a two-parameter histogram. It is interest-
ter profile (FS vs. SS), as is customary in flow ing to note that three distinct cell clusters can
cytometry. A rectilinear region was then estab- be distinguished on the basis of the correlated
lished to exclude dead cells, debris, and cell expression of CD62L and CD25 expression.
aggregates from further analysis. This process One strategy is to “color-event” the events of
is referred to as gating. The FS-versus-SS pro- interest. As shown in Figure 18.8.1D, two re-
file of dead cells, as determined by PI uptake, gions have been set around the cells that have
indicated that these cells were located below down-regulated their expression of CD62L,
the gating region (data not shown). In gen- one population expresses CD25 (color-evented
Immunotoxicology
eral, the authors find that the viability of their red), and another does not express CD25
18.8.19
Current Protocols in Toxicology Supplement 23
Figure 18.8.1 Legend at right.

Immune Cell
Phenotyping
Using Flow
Cytometry

18.8.20
Supplement 23 Current Protocols in Toxicology
(color-evented blue). The cells that have re- clusters. In addition, this new approach also
tained their CD62L expression have been makes it easier to assess compensation. Both
color-evented gray. In this way, the expression FlowJo (Tree Star) and WinList (Verity Soft-
of all their markers can be easily visualized for ware House) offer the option to display flow
each population in all the histograms. For in- cytometric data using the new Logicle axis.
stance, the red population also expresses high
levels of CD28 and elevated levels of CD4. In Statistics
contrast, the blue population expresses inter- The data shown in Figure 18.8.1 represent
mediate levels of CD28 and CD4. By defini- the response of one individual animal. In most
tion, both the red and blue cells are consid- immunotoxicological studies, the authors use
ered activated T cells since they have down- four to six animals per treatment group. In or-
regulated their expression of CD62L; however, der to determine a treatment effect, data de-
the red cells are further along in the activa- rived from the treatment group must be com-
tion pathway since they have also up-regulated pared with data from the vehicle control group.
their expression of CD25, which occurs only The authors will often include a group of naive
after several cell divisions. The gray popula- animals (N = 2 to 3) in order to monitor
tion represents CD4+ cells that have not be- the antigen-induced response of the control
come activated by day two of the GvH re- animals.
sponse. In general, the authors prefer to present flow
There is a new approach in visualizing flow cytometric data in the form of histograms and
cytometric data, i.e., changing the traditional use statistics to state the level of confidence in
four-decade logarithmic scale for immunoflu- the accuracy of each result. Flow cytometric
orescent data displayed in two-parameter his- data are usually presented as the frequency of
tograms to a new Logicle axis (Tung et al., any given population that expresses a particu-
2004). The plot axes have been redefined to al- lar marker and/or as a measure of the intensity
low the log-transformed scale to have a zero as of the fluorescent or light scatter signals. There
well as a negative region. There are several ad- are three measures commonly used to describe
vantages to this approach. Once data have been the central tendency when assessing fluores-
properly compensated, a large number of neg- cence intensity: mean, median, and mode. The
ative cells pile up in the first channel, against mean fluorescence can be significantly skewed
the axis, as these cells exhibit little to no flu- higher or lower by the presence of a few out-
orescence. Using the new Logicle scale, these liers. Therefore, when possible, one should use
negative cells can be visualized as distinct cell the median fluorescence of the positives, since

Figure 18.8.1 (at left) Five-color analysis of T cell activation in a graft versus host (GvH) model.
T cells purified from the spleens of C57Bl/6 mice (H-2b ; referred to as donors) were injected
into C57Bl/6 (H-2b ) x DBA/2 (H-2d ) F1 mice. These donor T cells recognize the H-2d antigens
contributed by the DBA/2 strain in the F1 host and become activated. Two days after the injec-
tion of donor T cells, the F1 mice were killed, and their spleens were processed into single-cell
suspensions. The spleen cells were stained simultaneously with MAbs to CD4, H-2Dd , CD28,
CD25, and CD62L. Cells were analyzed using an FC500 flow cytometer (Beckman Coulter). (A)
Viable cells were identified on the basis of their forward scatter (FS)–versus–side scatter (SS)
profiles. (B) CD4+ donor cells (elliptical region) were identified by their expression of CD4 and
their lack of expression of H2Dd . In this sample, the donor CD4+ cells represent 1.9% of the to-
tal spleen cell population. (C) Expression of the T cell activation molecules CD28, CD25, and
CD62L was assessed in single-parameter histograms by gating on 10,000 donor CD4+ cells. Cells
with positive staining for the specified MAbs are represented by the gray-filled histograms. Open
histograms represent negative staining associated with the isotype control antibodies. (D) Cor-
related expression of activation molecules on donor CD4+ cells as illustrated in two-parameter
histograms. Donor CD4+ cells that have up-regulated expression of CD25 are color-evented as
red events. Using color-eventing, a distinct cluster of donor CD4+ CD25+ cells that also express
low levels of CD62L and high levels of CD28 is readily identifiable. When the data are displayed
in a two-parameter histogram, the presence of at least three CD4+ subpopulations distinguished
by their differential expression of CD25 and CD62L is evident (far left histogram). It is interesting
to note that not all cells with low CD62L expression exhibit high CD25 expression. Also of interest
is that CD4+ CD25+ cells have up-regulated expression of CD4, as shown in (B). This black and
white facsimile of the figure is intended only as a placeholder; for full-color version of figure go to
http://www.interscience.wiley.com/c p/colorfigures.htm Immunotoxicology

18.8.21
Current Protocols in Toxicology Supplement 23
the median is generally a more robust estimate sions; and (4) the use of a verification con-
of central tendency than the mean. trol to establish the accuracy of the results
The precision in cell counting, or the ex- yielded by the system. Once the instrument
tent to which replicate samples agree with each has been validated, there are several technical
other, is highly dependent on the number of issues that must be addressed, such as cellular
events sampled. The statistic which is most of- autofluorescence, FcR-mediated binding, and
ten used to measure precision is the coefficient the presence of dead cells, which can result
of variation (CV), which can be determined in the collection of inaccurate data and possi-
using the following simple equation: bly lead to data misinterpretation or erroneous
conclusions.

CV = [( N)/N] × 100% It is easy to obtain high precision in flow
cytometric data; this can be done by simply
where N is the number of events sampled and collecting data on a minimum of 10,000 cells
the square root of N represents the expected of interest. As an example, when assessing ac-
standard deviation. As the difference between tivation markers on CD4+ T cells, data should
two populations becomes smaller, better and be collected by gating on 10,000 of these cells.
better precision (i.e., a lower CV) is needed. The precision of a given measurement is usu-
For example, the CV for 25 cells is 20%; for ally characterized by the statistic CV, and for
100 cells, 10%; and for 10,000 cells, 1%. In 10,000 events, the expected CV would be 1%.
the authors’ laboratory, in order to attain high Thus, the assessment of 10,000 cells of inter-
precision, flow cytometric data are routinely est will help to ensure that small differences
collected on a minimum of 10,000 cells of in- between two populations may still be biologi-
terest, whether one is acquiring 10,000 viable cally significant.
cells or 10,000 antigen-specific T cells (such as It is only through these steps that one can
the CD4+ donor cells shown in Fig. 18.8.1B). be confident that the results obtained are accu-
rate, and this can ensure the validity of data
Anticipated Results interpretation. This is of utmost importance
The objective in flow cytometric analyses is when assessing treatment effects in toxicologi-
to resolve epitope-positive cells from epitope- cal studies. Treatment effects can be discerned
negative cells with a high degree of precision by changes in the relative frequency of a par-
and accuracy. Sometimes, it is necessary to se- ticular lineage-specific population (e.g., CD4+
lect a MAb that is labeled with a fluorochrome T cells, B cells, macrophages) or by changes
such as PE in order to maximize the resolu- in the relative frequency of a specific sub-
tion of positive cells from negative cells, es- set within these major leukocyte populations
pecially when assessing dim immunofluores- (e.g., activated CD4+ T cells, cytolytic T effec-
cence. However, there are several procedures tor cells). Sometimes it is necessary to report
that can be employed to ensure the collection changes in the MCF for a particular marker,
of accurate results. especially when all the cells of interest homo-
Proper setup will help to eliminate instru- geneously express that marker (e.g., CD11a,
mental error as a source of variation within an CD54). However, the bottom line in flow cyto-
experiment. The systematic process by which metric analysis is that each result is expected to
an instrument’s performance is validated is accurately reflect the cell population(s) being
known as quality control, and this process sampled. In this way, one can be assured that
should be conducted for every experiment. the data can be correctly interpreted and valid
There are several sequential quality control conclusions derived. In-depth discussions on
steps that should be followed (as discussed in the issues presented can be found in Stewart
Critical Parameters): (1) the use of standard- and Stewart (2001b) and Shapiro (2003).
ized beads, such as Flow-Check fluorospheres
(Beckman Coulter), to check laser alignment Time Considerations
as well as the fluidics and optics of the sys- The time required to process, stain, and
tem; (2) the use of standardized beads, such analyze samples is contingent on many fac-
as Flow-Set fluorospheres (Beckman Coulter), tors, including the number of animals in
for channel positioning (to ensure that changes the experiment and the number of stain-
observed in time-course studies are the result ing combinations. The time needed to pre-
Immune Cell of biological effects and not of changes in the pare single-cell suspensions from lymphoid
Phenotyping instrument); (3) the use of compensation con- tissues depends on the organ to be pro-
Using Flow trols to remove unwanted fluorescence from cessed (spleen, thymus, and/or lymph nodes).
Cytometry
fluorochromes with overlapping spectral emis- Spleen cell suspensions will require an extra
18.8.22
Supplement 23 Current Protocols in Toxicology
step in sample processing to remove RBCs, and treated with 2,3,7,8-tetrachlorodibenzo-p-
whereas thymus and lymph node cell suspen- dioxin. Int. Immunopharmacol. 3:553-570.
sions will not. The use of microtiter plates Food and Drug Administration. 2002. Guidance
will greatly expedite the staining of large for Industry. Immunotoxicology Evaluation of
Investigational New Drugs. http://www.fda.gov/
numbers of samples derived from lymphoid
cder/guidance/4945fnl.doc
organs. Even though blood samples require no
Funatake, C.J., Dearstyne, E.A., Steppan, L.B.,
further processing once the blood is collected,
Shepherd, D.M., Spanjaard, E.S., Marshak-
the staining process requires the use of test Rothstein, A., and Kerkvliet, N.I. 2004. Early
tubes or microcentrifuge tubes, making sam- consequences of 2,3,7,8-tetrachlorodibenzo-p-
ple manipulation more time-consuming. The dioxin exposure on the activation and survival
time required for cell staining is dependent on of antigen-specific T cells. Toxicol. Sci. 82:129-
142.
the number of cell samples to be stained and
the staining method employed (direct or in- Immunotoxicology Technical Committee, Interna-
tional Life Sciences Institute Health and Envi-
direct). In general, the direct staining method
ronmental Sciences Institute. 2001. Application
will take 10 min for FcR blocking and 10 min of flow cytometry to immunotoxicity testing:
for incubation with fluorochrome-conjugated Summary of a workshop. Toxicology 163:39-48.
MAbs, followed by 10 to 20 min for wash Kantor, A. and Roederer, M. 1997. FACS analysis
steps. Indirect staining will require an addi- of lymphocytes. In Handbook of Experimental
tional 20 min to incubate cells with the second- Immunology, 5th ed. (L.A. Herzenberg, D.M.
step reagent, followed by 10 to 20 min for wash Weir, L.A. Herzenberg, and C. Blackwell, eds.)
pp. 49.1–49.13. Blackwell Science, Cambridge,
steps. The time necessary to acquire flow cy-
UK.
tometric data will vary depending on cell con-
Kerkvliet, N.I. and Brauner, J.A. 1990. Flow cy-
centration and on the total number of events
tometric analysis of lymphocyte subpopulations
to be collected. For example, it should take in the spleen and thymus of mice exposed to
less than 1 min per sample to collect 10,000 an acute immunosuppressive dose of 2,3,7,8-
to 20,000 events when determining the vari- tetrachlorodibenzo-p-dioxin (TCDD). Environ.
ous lineage-specific populations in a sample Res. 52:146-154.
of spleen cells. However, it will definitely take Luster, M.I., Portier, C., Pait, D.G., Rosenthal, G.J.,
longer to collect 10,000 events when gating Germolec, D.R., Corsini, E., Blaylock, B.L.,
Pollock, P., Kouchi, Y., Craig, W., White, D.L.,
on a smaller population. Instrumental setup
Munson, A.E., and Comment, C.E. 1993. Risk
will take much longer for multicolor analy- assessment in immunotoxicology: II: Relation-
ses as compared with one-color analyses, as ships between immune and host resistance tests.
proper compensation will need to be estab- Fundam. Appl. Toxicol. 21:71-82.
lished in multicolor studies. The time involved Mitchell, K.A. and Lawrence, B.P. 2003. Exposure
in data analysis is contingent on the number of to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)
fluorochromes used in each MAb cocktail as renders influenza virus-specific CD8+ T cells
hyporesponsive to antigen. Toxicol. Sci. 74:74-
well as the number of samples processed. Data
84.
analysis can take as little as just a few min-
Molecular Probes. 2004a. Zenon technology: Ver-
utes per sample for samples stained with one
satile reagents for immunolabeling. In Hand-
fluorochrome-conjugated MAb and as many book of Fluorescent Probes and Research Prod-
as several hours for samples stained with five ucts, 9th ed. http://www.probes.com/handbook/
different colors. sections/0703.html
Molecular Probes. 2004b. Alexa Fluor dyes: Sim-
Acknowledgement ply the best. In Handbook of Fluorescent Probes
The authors would like to acknowledge and Research Products, 9th ed. http://www.
Castle Funatake for her valuable editorial as- probes.com/handbook/sections/0103.html
sistance. This unit was supported by the Cell Nygaard, U.C. and Lovik, M. 2002. Blood and
and Tissue Analysis Facilities and Services spleen lymphocytes as targets for immunotoxic
effects in the rat: A comparison. Toxicology
Core of the Environmental Health Sciences 174:153-161.
Center, Oregon State University, grant num-
Oughton, J.A., Pereira, C.B., DeKrey, G.K.,
ber P30 ES00210, National Institute of Envi- Collier, J.M., Frank, A.A., and Kerkvliet, N.I.
ronmental Health Sciences, National Institutes 1995. Phenotypic analysis of spleen, thymus,
of Health. and peripheral blood cells in aged C57Bl6
mice following long-term exposure to 2,3,7,8-
tetrachlorodibenzo-p-dioxin. Fundam. Appl.
Literature Cited Toxicol. 25:60-69.
Choi, J.-Y., Oughton, J.A., and Kerkvliet, N.I.
2003. Functional alterations in CD11b+ Gr-1+ Riedy, M.C., Muirhead, K.A., Jensen, C.P., and
cells in mice injected with allogeneic tumor cells Stewart, C.C. 1991. Use of a photolabeling Immunotoxicology

18.8.23
Current Protocols in Toxicology Supplement 23
technique to identify nonviable cells in fixed ho- Stewart, C.C. and Stewart, S.J. 2001b. Multiparam-
mologous or heterologous cell populations. Cy- eter data acquisition and analysis of leukocytes
tometry 12:133-139. by flow cytometry. In Methods in Cell Biol-
Roederer, M. 1997. Conjugation of Monoclonal ogy, Vol. 63 (Z. Darzynkiewicz, H.A. Crissman,
Antibodies. http://www.drmr.com/abcon/index. and J.P. Robinson, eds.) pp. 289-312. Academic
html Press, San Diego.

Roederer, M. 2000. Compensation in Flow Cy- Tung, J.W., Parks, D.R., Moore, W.A., and Herzen-
tometry: An Informal Perspective. http://www. berg, L.A. 2004. New approaches to fluores-
drmr.com/compensation/index.html cence compensation and visualization of FACS
data. Clin. Immunol. 110:277-283.
Shapiro, H.M. 2003. Practical Flow Cytometry, 4th
ed. John Wiley & Sons, Hoboken, N.J. Vandebriel, R.J., Spiekstra, S.W., Hudspith, B.N.,
Meredith, C., and Van Loveren, H. 1999. In vitro
Shepherd, D.M., Dearstyne, E.A., and Kerkvliet, exposure effects of cyclosporin A and bis(tri-n-
N.I. 2000. The effects of TCDD on the acti- butyltin)oxide on lymphocyte proliferation, cy-
vation of ovalbumin (OVA)-specific DO11.10 tokine (receptor) mRNA expression, and cell
transgenic CD4(+) T cells in adoptively trans- surface marker expression in rat thymocytes and
ferred mice. Toxicol. Sci. 56:340-350. splenocytes. Toxicology 135:49-66.
Stewart, C.C. and Stewart, S.J. 2001a. Cell prepa-
ration for the identification of leukocytes. In
Methods in Cell Biology, Vol. 63 (Z. Contributed by Julie A. Oughton and
Darzynkiewicz, H.A. Crissman, and J.P. Robin- Nancy I. Kerkvliet
son, eds.) pp. 217-251. Academic Press, San Oregon State University
Diego.
Corvallis, Oregon

Immune Cell
Phenotyping
Using Flow
Cytometry

18.8.24
Supplement 23 Current Protocols in Toxicology

Potrebbero piacerti anche