Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Use of the Intracellular Fluorescent Dye UNIT 4.

9
CFSE to Monitor Lymphocyte Migration
and Proliferation
Christopher R. Parish,1 Megan H. Glidden,1 Ben J. C. Quah,1 and Hilary S.
Warren1
1
Australian National University, Canberra, Australia

ABSTRACT
The stable incorporation of the intracellular fluorescent dye 5-(and -6)-carboxyfluorescein
diacetate succinimidyl ester (CFSE) into cells provides a powerful tool to monitor cell
migration, and to quantify cell division, because of the sequential decrease in fluorescent
labeling in daughter cells. CFSE-labeled lymphocytes have been used to analyze the
relationship between cell division and differentiation of cell function, and cell prolifera-
tion versus apoptosis, both in vivo and in vitro, and have allowed analysis of the site of
response to antigens in vivo. Curr. Protoc. Immunol. 84:4.9.1-4.9.13.  C 2009 by John

Wiley & Sons, Inc.


Keywords: CFSE r cell division r cell tracking r lymphocyte migration r
lymphocyte positioning

INTRODUCTION
The stable incorporation of the intracellular fluorescent dye 5-(and -6)-carboxyfluorescein
diacetate succinimidyl ester (CFSE) into lymphocytes (Basic Protocol) is a powerful tool
to monitor lymphocyte migration in vivo, and to quantitatively analyze cell division both
in vivo and in vitro (Support Protocol). The stability of CFSE labeling allows monitoring
of lymphocytes over a period of months in vivo. Cell division results in sequential halving
of fluorescence, and up to 8 divisions can be monitored before the fluorescence is de-
creased to the background fluorescence of unstained cells. The relationship between cell
division and cell function is readily measured at the time of analysis by using a cell func-
tion marker (cell surface or intracellular protein) labeled with an alternate fluorochrome.
Similarly, T and B lymphocyte subsets and NK cells can be individually analyzed for
cell division in a complex population by using appropriate cell surface markers. CFSE
remains associated with apoptotic cells for several days, and these can be analyzed to-
gether with live cells by appropriate electronic gating (by size and granularity) on the
flow cytometer. Since halving of fluorescence occurs in daughter cells, by calculating
the proportion of cells in each division peak and dividing by the expected progeny at
those divisions (2, 4, 8, 16, etc.), the number of cells that have entered division can be
calculated. This gives a precursor frequency estimate of responding cells in the cultures.

CFSE LABELING OF LYMPHOCYTES BASIC


PROTOCOL
This protocol describes methods for labeling high or low numbers of lymphocytes with
CFSE. Steps are provided to use CFSE-labeled cells in cell transfer studies in vivo or
as cells to be cultured in vitro. If analysis of cell migration is a goal of the experiment,
specific guidelines for positioning of CFSE-labeled lymphocytes in lymphoid organs
or other tissues are provided in this protocol. A Support Protocol for flow cytometric
analysis of CFSE-labeled cells follows. In Vivo Assays
for Lymphocyte
Function

Current Protocols in Immunology 4.9.1-4.9.13, February 2009 4.9.1


Published online February 2009 in Wiley Interscience (www.interscience.wiley.com).
DOI: 10.1002/0471142735.im0409s84 Supplement 84
Copyright C 2009 John Wiley & Sons, Inc.
Materials
Experimental animals or human peripheral blood or cultured lymphocytes
Phosphate-buffered saline (PBS; APPENDIX 2A), pH 7.4
Hanks’ balanced salt solution (HBSS), pH 7.4 (APPENDIX 2A)
PBS (APPENDIX 2A) containing 5% (v/v) heat-inactivated FBS
5 mM CFSE stock solution (see recipe)
Antigens and mitogens of interest
0.5 mM disodium EDTA in PBS (APPENDIX 2A)
1-ml syringes
25-G needles
Fluorescence microscope with filters for fluorescein
Razor blade
Additional reagents and equipment for removal of mouse lymphoid organs
(UNIT 1.9), preparation of mononuclear cell suspensions (UNIT 3.1), and isolation of
peripheral blood mononuclear cells (UNIT 7.1), immunohistochemistry (UNIT 21.4),
culturing mouse (UNITS 3.10 & 3.12), or human (UNITS 7.10 & 7.11) lymphocytes, and
counting cells using a hemacytometer (APPENDIX 3A)
Label lymphocytes with CFSE
1a. For high cell numbers: Prepare lymphocytes using the techniques described in UNIT 1.9
(for mice; removal of lymphoid organs), UNIT 3.1 (preparation of cell suspensions),
and UNIT 7.1 (preparation of PBMC), at a concentration of 50 × 106 cells/ml in
either PBS (without serum) for human PBMC or HBSS (without serum) for mouse
lymphocytes.
1b. For low cell numbers: Resuspend freshly isolated lymphocytes in PBS containing
5% FBS at concentrations from 0.5 × 106 cells/ml to 10 × 106 cells/ml.
At low cell concentrations it is absolutely essential that there be protein present to buffer
the toxic effect of CFSE.
Cultured lymphocytes that are quiescent at the end of primary culture are labeled directly
in their culture medium (containing 10% FBS) after equilibrating to room temperature.
2. Dilute the stock 5 mM CFSE solution 1/100 in PBS (to give a 50 μM solution). Add
110 μl of this solution per milliliter of cells (to give a final concentration of 5 μM),
and mix rapidly. After 5 min at room temperature add 10 vol of PBS containing 5%
FBS, centrifuge cells 5 min at 300 × g, 20◦ C, and remove the supernatant. Wash three
times, each time by resuspending in 10 vol PBS containing 5% FBS, centrifuging
5 min at 300 × g, 20◦ C, and removing the supernatant.
Labeling with CFSE occurs rapidly, and it is essential that CFSE be dispersed as evenly
and quickly as possible so that cells are uniformly labeled. One strategy to achieve this
is to add the cell suspension into the bottom of a 10-ml plastic tube, then, while holding
the tube almost horizontally, add the CFSE solution to a nonwetted portion of the plastic
at the top of the tube. The tube is then capped while still in the near-horizontal position,
and then rapidly inverted several times to mix the lymphocytes and CFSE solution. An
alternate strategy is to predilute the CFSE to 10 μM and add an equal volume to the
cell suspension while vortexing. If this strategy is used for high cell numbers, prepare the
lymphocytes at 100 × 106 cells/ml instead of 50 × 106 cells/ml.
When labeling cultured lymphocytes it is best to add CFSE directly into the existing culture
medium without prior centrifugation. When cultured cells are centrifuged they form small
aggregates such that individual cells are not exposed equally to CFSE. After labeling
Use of CFSE to cultured lymphocytes with CFSE, the cells are washed in PBS and then incubated for
Monitor
Lymphocyte 5 min in 0.5 mM EDTA in PBS to dissociate any aggregates, and washed once more in
Migration and PBS before resuspending in culture medium for restimulation in culture.
Proliferation

4.9.2
Supplement 84 Current Protocols in Immunology
CFSE staining of lymphocytes cannot be measured directly after labeling because of the
extremely high fluorescence. The majority of CFSE initially taken up by the cells is not
stably incorporated and is lost within the first 24 hr after labeling.
Perform in vivo transfer of CFSE-labeled lymphocytes
3. Resuspend CFSE-labeled lymphocytes in tissue culture medium lacking added pro-
tein (no serum), and, using a 1-ml syringe equipped with a 25-G needle, inject
intravenously (i.v.) into recipient animals. In the case of mice, inject into the lateral
tail vein, with 1 × 106 to 40 × 106 cells being injected into each recipient mouse in
a volume of 0.1 to 0.2 ml.
There is a linear increase in the number of CFSE-labeled cells entering mouse lymphoid
organs with the transfer of up to 50 × 106 cells, but when greater numbers of cells are
transferred the system appears to become saturated.
If extensive proliferation of the CFSE-labeled population is anticipated, it is essential
that the cells have an independent marker for detection from host cells that does not
decrease with cell division. This is most commonly satisfied by using cells that have a
CD45 allotypic difference from host animals, e.g., injection of CFSE-labeled CD45.1+
cells into CD45.2+ host animals (see Critical Parameters and Fig. 4.9.1).
If, on the other hand, in vivo migration of lymphocytes is being investigated under condi-
tions of minimal cell division, it is possible to independently track two different lymphocyte
populations in the same animal by labeling the cells to different fluorescence intensities
with CFSE (Lyons, 1999). One population is labeled with 5 μM CFSE (see step 1a or 1b
above) and the other population with one-quarter (1.25 μM) or one-sixteenth (0.312 μM)
the normal CFSE-labeling concentration.
If one plans to examine the CFSE-labeled cells <24 hr after they have been transferred
into recipient animals, in order to avoid off-scale fluorescence intensities on the flow
cytometer, the lymphocytes should be labeled with one-quarter (1.25 μM) or one-eighth
(0.625 μM) the normal CFSE-labeling concentration (see step 1 or 2 above).

4. Detect the positioning of CFSE-labeled lymphocytes within lymphoid organs and


other tissues by fluorescence microscopy with filter settings for fluorescein, or
by immunohistochemistry (UNIT 21.4) using fluorescein-specific antibodies (Garton
and Schoenwolf, 1996; Graziano et al., 1998). For fluorescence microscopy de-
tection of CFSE-labeled cells, remove organs from animals, cut ∼3-mm sections
of the organs with a razor blade, and place the sections on microscope slides for
examination.
CFSE fluorescence is rapidly quenched when tissue sections are viewed by fluorescence
microscopy and is totally quenched when the sections are treated with conventional his-
tological stains. For short-term positioning studies of relatively low resolution, the DNA-
intercalating dye H33342 is recommended as a highly fluorescent, quenching-resistant
dye for labeling lymphocytes (Parish, 1999).
If high-resolution positioning studies are required, immunohistochemical detection of
CFSE-labeled cells in tissue sections is recommended.

Culture lymphocytes
5. Resuspend CFSE-labeled lymphocytes in culture medium and stimulate in vitro with
antigens or mitogens of interest.
Procedures for culturing mouse and human lymphocytes are detailed in UNITS 3.10 & 3.12,
and UNITS 7.10 & 7.11, respectively.

Harvest cells
6a. For in vivo harvesting: Collect lymphoid organs, and make single-cell suspensions
(UNITS 1.9 & 3.1). In Vivo Assays
for Lymphocyte
Function

4.9.3
Current Protocols in Immunology Supplement 84
A
1000 104
12000 lymphocytes 1200 CD8α + cells
800

CD8α-APC-A
103

# Cells
9000 900
SSC-A

600

# Cells
6000 102 600
400
1
200 3000 10 300

0 0 100 0 0
0 200 400 600 800 1000 100 101 102 103 104 10 101 102 103 104 100 101 102 103 104
FSC-A CFSE-A B220-PerCP-Cy5-5-A CFSE-A
104 12000 104
Hoechst 33258-A

live cells Vα-2+ cells


3
100
3 10
10 9000

Vα-2-PE-A
80
# Cells

# Cells
102 6000 10 2 60
92.5% 12.2% 40
101 3000 101
20

10 0 0 100 0
0 200 400 600 800 1000 100 101 102 103 104 100 101 102 103 104 100 101 102 103 104
FSC-W CFSE-A CFSE-A CFSE-A
CD45.1-PE-Cy7-A 104 40 CD45.1+
3
cells
10 30

# Cells
102 20
31.4%
101 10

100 0 0
10 101 102 103 104 100 101 102 103 104
CFSE-A CFSE-A
B
CFSE-OT-I CD8+ cells CFSE-OT-I CD8+ cells Unlabeled-CD8+ cells
+ OVA + PBS + 20 μg OVA

2 μg 20 μg 200 μg
100 100 100
80 80 80
% of Max

60 60 60
40 40 40
20 20 20
0 0 0
100 101 102 103 104 100 101 102 103 104 100 101 102 103 104
CFSE-A

Figure 4.9.1 Progressive gating strategy for detection of maximum T cell divisions in vivo. OVA-specific Vα2+ , CD8α+
T cells were purified from OT-I mice (bred on a CD45.1 background) and labeled with CFSE. T cells (5 × 106 ) were
then intravenously injected into C57BL/6J mice (which have a CD45.2 background). At a time point 2 hr later, mice were
intravenously injected with either 2, 20, or 200 μg of OVA (in PBS) or PBS alone as a control. After 3 days, splenocytes
were harvested, labeled with Hoechst 33258 (at 1 μg/ml for dead cell discrimination) and APC-conjugated anti-CD8α,
PerCP-Cy5.5-conjugated anti-B220, PE-conjugated anti-Vα2, and PE-Cy7 conjugated anti-CD45.1 monoclonal antibodies,
and 1 × 106 cells were collected on an LSR-II (BD biosciences) for analysis. (A) Progressive gating on subpopulations
of splenocytes reveals higher resolution of CFSE profiles of transferred cells (with CD8α+ , CD45.1+ viable cells giving
the highest resolution). (B) Detection of division of transferred CFSE-labeled OT-I CD8+ T cells to increasing amounts of
antigen (OVA).

4.9.4
Supplement 84 Current Protocols in Immunology
6b. For in vitro harvesting: Harvest cells from culture, wash once in 3 ml PBS, resuspend
in 2 ml 0.5 mM EDTA/PBS, and incubate for 5 min at 37◦ C to dissociate aggregates.
Centrifuge cells 5 min at 30 × g, 20◦ C, resuspend in PBS containing 5% FBS,
and transfer to tubes suitable for use with the flow cytometer. If the cells are to be
counted manually with a hemacytometer (APPENDIX 3A), retain a small sample on ice
in a separate tube or V-well plate.
Cells for analysis on the flow cytometer are not fixed with paraformaldehyde, but are kept
on ice until analyzed.
For multiple cultures, it is convenient to use 96-well V-well plates. For cultures harvested
from 96-well flat-well plates, an aliquot of supernatant from each of the wells is first
discarded so that the triplicate cultures can be combined into 1 well of the 96-V-well
plate. Subsequent washing and treatment with EDTA then occurs in 150-μl volumes in
these wells.
The plates are centrifuged 2 min at 300 × g, 4◦ C in a plate holder. The supernatant is
removed using a microtiter pipettor by following the meniscus down while aspirating until
the tip reaches the intersection of the vertical wall and the top of the V-base of the well.
The pellet is mixed by agitating the plate over a vortex mixer prior to adding the next wash
solution with a multipipettor.

ANALYSIS OF CFSE-LABELED CELLS BY FLOW CYTOMETRY SUPPORT


PROTOCOL
This protocol summarizes the steps used to analyze CFSE-labeled cells in order to
quantify cell division. The proportion of cells in the individual CFSE peaks can be
determined manually or by applying software that deconvolutes the peaks. Guidelines
for either approach are provided in this protocol. The Critical Parameters section of
this unit describes important parameters related to the analysis of CFSE-labeled cells
that have been transferred in vivo or cultured in vitro following labeling. Additional
information about flow cytometric analysis of cells can be found in Chapter 5. In this
protocol, the choice of which other labeling procedures are to be used (e.g., cell surface
staining, intracellular staining for cytokines, identification of apoptotic cells) depends on
the goals of the experiment at hand.

Materials
CFSE-labeled cells (see Basic Protocol)
Analytical flow cytometer capable of 3-color fluorescence
Cell sorting flow cytometer (for some applications)
Additional reagents and equipment for cell surface staining of lymphocytes
(UNIT5.3), intracellular cytokine staining (UNIT 6.24), analytical flow cytometry
(UNIT 5.4), flow cytometric analysis of apoptosis (UNIT 3.17), and cell sorting flow
cytometry (UNIT 5.1)
1. Perform flow cytometric staining steps suited to the experiment being performed
(e.g., cell surface staining, UNIT 5.3; intracellular cytokine staining, UNIT 6.24; Hoechst
33258 staining or propidium iodide staining for measurement of apoptosis, UNIT 3.17).
Perform flow cytometric analysis (UNIT 5.4).
CFSE is a fluorescein-based dye. Labeling lymphocytes for cell surface staining or in-
tracellularly for cytokines requires the use of an alternative fluorochrome such as PE,
PE-Cy7, PerCp-Cy5.5, APC, APC-AlexaFluor 750 etc. (see Table 6.21.2).

For software-based calculations


2a. Deconvolute CFSE peaks using appropriate software.
Several software programs, such as Profit for Macintosh (Quantumsoft), Peakfit for PC In Vivo Assays
(SPSS Sciences), and ModFit (Verity Software House; see Internet Resources) are avail- for Lymphocyte
able for deconvoluting CFSE peaks. Programs such as Flowjo (Tree Star; see Internet Function

4.9.5
Current Protocols in Immunology Supplement 84
Resources) can also be used on both a Macintosh and PC for analyzing CFSE profiles. For
more detailed instructions on analysis of CFSE profiles using software refer to Hawkins
et al. (2007).

For manual calculations


2b. Record the geometric mean fluorescence of the control CFSE-stained cells and the
unstained cells of both the control and stimulated cultures.
For example, one might have a geometric mean fluorescence of 600 for CFSE staining
of control unstimulated cells compared to an autofluorescence control of 2 for stimulated
unstained cells.
Note that the autofluorescence value of the unstained cells is greater for the dividing
compared to the control nondividing cells.

3b. Subtract the geometric mean fluorescence of the appropriate control from that of the
CFSE control cells (e.g., 600 − 2 = 598).
4b. Convert this value to its base 10 logarithm (in the above case, 2.776).
5b. Determine the geometric mean fluorescence of daughter populations. As these occur
at 1/2, 1/4, etc. of the undivided peak, subtract 0.3 log10 units.
Thus, continuing with the above example, the successive peaks are at 2.476 (division 1),
2.176 (division 2), 1.876 (division 3), 1.576 (division 4), 1.276 (division 5), 0.976 (division
6), and 0.676 (division 7).

6b. Determine the boundaries of the peaks.


The boundaries are midway between successive peaks and are therefore 0.15 log10 units
each side of the peak. Therefore, the lower (left) boundary for each peak is as follows:
2.626 (undivided control cells), 2.326 (division 1), 2.026 (division 2), 1.726 (division 3),
1.426 (division 4), 1.126 (division 5), 0.826 (division 6), and 0.526 (division 7). Next, the
antilog of these figures is determined: 423, 212, 106, 53.2, 26.7, 13.4, 6.7, and 3.4. To
these values are added the geometric mean autofluorescence (2 in this example) to give
425, 214, 108, 55.2, 28.7, 15.4, 8.7, and 5.4.
7b. Apply these markers using a flow cytometry program (such as Cell Quest, Becton
Dickinson) and determine the % of cells in each peak.
8b. Determine the actual number of cells in each peak from the manual counts or counting
bead analysis.
9b. Divide the cell numbers in each peak by the expected progeny for those divisions.
Thus, divide by 2 for 1 division, divide by 4 for 2 divisions, divide by 8 for 3 divisions,
etc.
The total of these numbers gives the number of progenitor cells and can be compared with
the number of cells in control cultures.
In some cases applying the above calculations gives markers that are not aligned exactly
over the peaks. When the calculations are redone using division 1 as a reference peak,
the markers then align correctly. It appears that during the transition from undivided to
the first division there is some loss of incorporated CFSE. This appears to occur with
some cell types more than others. The example given in Figure 4.9.2 for human peripheral
blood lymphocytes responding to phytohemagglutinin is a case in point. You will note that
the geometric mean for the undivided cells is 763, which is more than twice that of the
peak in division 1. The remainder of the peaks are close to the expected halving with each
division.
Use of CFSE to This quantitative analysis of cell division giving the number of progenitor cells provides
Monitor
Lymphocyte useful information. First, the lymphocyte population may be stimulated to divide and
Migration and to undergo activation-induced cell death. Dead cells retain the CFSE dye, and thus the
Proliferation division number in which death of a cell occurred can be readily determined. Dead cells

4.9.6
Supplement 84 Current Protocols in Immunology
M8
M7
M6
M5
M4
M3
M2
M1

0
100 101 102 103 104

Division % at
Marker Left Right Geometric number Progeny each
(M) boundary boundary mean % (i) 2i Progenitors division
1 437 9646 763.13 18.10 0 1 18.10 70.73
2 220 437 310.07 4.24 1 2 2.12 8.28
3 111 220 153.92 5.66 2 4 1.415 5.53
4 58.29 111 77.57 11.80 3 8 1.475 5.76
5 29.43 58.29 40.89 23.47 4 16 1.467 5.73
6 16.55 29.43 22.44 26.22 5 32 0.819 3.20
7 9.31 16.55 13.53 12.05 6 64 0.188 0.73
8 1 9.31 7.63 0.74 7 128 0.006 0.02

Figure 4.9.2 CFSE profiles for PBMC labeled at 1 × 106 /ml in PBS containing 5% FBS with 5 μm
CFSE for 5 min and cultured with 5 μM phytohemagglutinin for 4 days. The unfilled peak on the
right at 7.6 × 106 fluorescence units corresponds to the control unstimulated CFSE-labeled cells.
The unfilled peak on the left at 2 × 100 fluorescence units corresponds to the autofluorescence of
unlabeled cells. The calculated marker positions are shown: M1 (undivided), M2 (1 division), M3
(2 divisions), M4 (3 divisions), M5 (4 divisions), M6 (5 divisions), and M7 (6 divisions).

can be identified by forward and side scatter or by propidium iodide uptake. Comparison
of the number of progenitor cells in the stimulated and control cultures will show if
significant cell death has occurred. Second, the percentage of progenitor cells in divisions
1 through 7 compared to divisions 0 through 7 is an estimate of the precursor frequency,
i.e., the proportion of the lymphocyte population with reactivity to a particular antigen or
mitogen.
In the experiment shown in Figure 4.9.2, 29.63% of cells have entered division. The
limitation using this technique for the determination of precursor frequencies is the number
of divisions that occur before the fluorescence of the dividing cells merges with the
autofluorescence of unstimulated cells.

REAGENTS AND SOLUTIONS


Use deionized, distilled water in all recipes and protocol steps. For common stock solutions, see
APPENDIX 2A; for suppliers, see APPENDIX 5.

CFSE solution
Dissolve 5- (and 6-) carboxyfluorescein diacetate succinimidyl ester (CFSE; Molec-
ular Probes) in dimethyl sulfoxide (DMSO) at a final concentration of 5 mM. Store
in suitably sized aliquots up to at least 12 months at −20◦ C.
The stock solution can be refrozen provided this is done as soon as possible after thawing.
In Vivo Assays
Prepare the substock of diluted CFSE just before use. Do not refreeze the substock. for Lymphocyte
Function

4.9.7
Current Protocols in Immunology Supplement 84
COMMENTARY
Background Information Thus, CFSE is an extremely valuable reagent
Unlike most other cells in vertebrates, the for immunological studies, as lymphocytes la-
cells of the immune system have the remark- beled with CFSE can be simultaneously mon-
able ability to continually migrate throughout itored for their proliferation status (either in
the body and position themselves in specific vitro or in vivo) and their migration and posi-
locations within tissues, particularly within tioning behavior in vivo.
lymphoid organs. In addition, following con- There is some confusion in the literature
tact with antigen, there is a rapid clonal expan- regarding the nomenclature for CFSE. The
sion of antigen-specific T and B lymphocytes, compound used to label cells is carboxyflu-
with the migration and positioning behavior of orescein diacetate succinimidyl ester, and thus
the proliferating lymphocytes often being very a more appropriate acronym would be CFDA-
different from their precursors. SE rather than CFSE. Unfortunately, when the
In order to fully understand a functioning authors first obtained the dye from Molecular
immune system, techniques are required that Probes in the late 1980s, it was listed in the
can simultaneously follow lymphocyte prolif- catalog as CFSE and this acronym was used
eration, lymphocyte migration into different in the first publications from the authors’ lab-
lymphoid organs, and the positioning pattern oratory reporting the use of the dye. In more
of the lymphocytes within lymphoid organs. recent Molecular Probes catalogs, the dye is
Early studies employed radioactive markers listed as CFDA-SE, but in order to be con-
to measure lymphocyte proliferation and fol- sistent and avoid confusion, the authors have
low lymphocyte migration. [3 H]thymidine has continued to use the CFSE acronym in all of
been widely used to follow lymphocyte pro- their publications.
liferation, both in vitro (UNIT 7.10) and in vivo. To fully appreciate the use of CFSE as
However, this approach suffers from the disad- a dye for studying lymphocyte proliferation
vantage that it is difficult to assess the subpop- and migration, it is important to understand
ulation of lymphocytes that are proliferating, the molecular basis of cell labeling by CFSE.
tedious autoradiography procedures being re- Figure 4.9.3 depicts the various stages in the la-
quired to enumerate the proliferating cells. beling of cells with CFSE. Carboxyfluorescein
Furthermore, the procedure only measures diacetate succinimidyl ester (CFDA-SE, see
those cells that are in S phase at the time of Fig. 4.9.3), because of two acetate side chains,
[3 H]thymidine addition. Bromodeoxyuridine is nonfluorescent and highly membrane per-
(UNIT 4.7) is an excellent reagent for measur- meant. Thus, this compound is rapidly taken
ing lymphocyte turnover in vivo, but yields up by cells, although due to its lipophilic nature
limited migration and positioning information. CFDA-SE also freely exits from cells. Once
The γ -emitting isotope 51 Cr has been used for inside cells, intracellular esterases remove the
many years to follow the distribution pattern acetate groups. The resultant carboxyfluores-
of injected lymphocytes in vivo but has a num- cein succinimidyl ester (CFSE, see Fig. 4.9.3)
ber of technical limitations (Ford, 1978), and is highly fluorescent and, due to its reduced
yields no information about the phenotype, lipophilicity, it exits from cells at a much
proliferation status, and positioning pattern of slower rate. This slower exit rate also pro-
the injected cells. longs the time available for CFSE to covalently
During the last 25 years, at least 14 differ- couple to intracellular molecules. The succin-
ent fluorescent dyes have been used to moni- imidyl side chain of CFSE is highly reactive
tor lymphocyte migration (Parish, 1999), with with amino groups, resulting in carboxyfluo-
the fluorescein-based dye CFSE emerging as rescein (CF) being coupled, via a very stable
the most versatile fluorescent dye. CFSE is a amide bond, to the amino groups of intracel-
membrane-permeant dye that can very stably lular molecules. In some cases, CF is coupled
label cells by covalently coupling to intracellu- to molecules to form conjugates (CFR1) that
lar molecules. CFSE was originally developed are rapidly degraded or still exit through the
as a long-term tracking dye for lymphocyte plasma membrane, whereas in other cases the
migration studies (Weston and Parish, 1990). CF conjugates (CFR2) are stable and unable
It soon became apparent that a major advan- to exit from the cell (see Fig. 4.9.3). It is these
Use of CFSE to tage of CFSE is that it can be used to monitor long-lived conjugates that allow stable label-
Monitor lymphocyte proliferation, due to the progres- ing of cells with CFSE to be achieved.
Lymphocyte
Migration and sive halving of CFSE fluorescence in cells fol- Since the mid-1990s many laboratories
Proliferation lowing cell division (Lyons and Parish, 1994). have used CFSE to track lymphocyte cell

4.9.8
Supplement 84 Current Protocols in Immunology
O CFDASE O
CH C O O O C CH
3 3
O
O 6
O cell exterior
NO C5
O O
cell membrane

HO O OH

O O R1 NH O CFR1
CH3 C O O O C CH3 HO O OH O
6
O R1 NH C 5
O
O O 6 O O
O 6 esterases
NOC 5 NOC5
O O O O HO O OH
CFDASE CFSE R2 NH
O CFR2
O
cytosol 6
R2 NH C 5
O

Figure 4.9.3 A schematic representation of the various stages in the labeling of cells with
carboxyfluorescein diacetate succinimidyl ester. For details see text. The size of the arrows in the
figure is proportional to the rate of diffusion of the different molecules through the cell membrane.
The carbonyl group located between positions 5 and 6 on the benzene ring is to indicate that the
dye is a mixture of 5- and 6-carbonyl structural isomers. Figure reproduced from Parish (1999)
with kind permission of the publishers (Blackwell Science Asia).

division both in vitro and in vivo (reviewed 1997; Sukkar et al., 2004). CFSE labeling has
in Hasbold et al., 1999; Parish, 1999; Warren, also been reported for tumor cell tracking (von
1999; Lyons, 2000; Quah et al., 2007). An im- Horsten et al., 2000), lysis of leukemic precur-
portant feature of these studies is that many sor cells (Jedema et al., 2004), nuclei label-
biological processes in lymphocytes, such as ing (Hasbold and Hodgkin, 2000), hematopoi-
T cell cytokine production, B cell Ig isotype etic stem cell proliferation (Oostendorp et al.,
switching, T cell apoptosis, and cell surface 2000), and the in vitro and in vivo assessment
molecule expression have been shown to be of cytotoxicity (Marzo et al., 2000; Hermans
very division-dependent. A particularly pow- et al., 2004; Stambas et al., 2007).
erful application of CFSE-labeling has been
to monitor T lymphocyte proliferation in re- Critical Parameters
sponse to a range of co-stimulatory signals CFSE is toxic to cells at high concentra-
(Gett and Hodgkin, 2000). Based on their ex- tions, and it is therefore essential to deter-
perimental data, Gett and Hodgkin derived an mine the optimum labeling conditions that
elegant mathematical model for the assess- give good fluorescence and preserve normal
ment of the effect of different signals on T cell function (Quah et al., 2007). For human pe-
clonal expansion. The three key variables that ripheral blood lymphocytes at concentrations
emerged as determining the magnitude of the T below 10 × 106 /ml, reliable labeling is best
cell proliferative response were the time to first achieved with the cells resuspended in PBS
division, the subsequent rate of division, and containing 5% FBS and using CFSE at a final
the rate of apoptosis, with different cytokines concentration of 5 μM. These conditions give
and co-stimulatory molecules having unique a fluorescence intensity between the second
effects on these three parameters. Although the and third decade when measured at 4 days.
majority of studies have used CFSE to follow Labeling exceeding the third decade gives im-
lymphocyte division, the proliferation of cell paired functional responses. This is illustrated
types as diverse as fibroblasts, airway smooth in Figure 4.9.4, showing the relationship, af- In Vivo Assays
muscle cells, and bacteria has been measured ter 4 days culture, between the amount of for Lymphocyte
using CFSE (Khil et al., 1997; Ueckert et al., CFSE in control unstimulated human PBMC Function

4.9.9
Current Protocols in Immunology Supplement 84
50

40

% of cells in division
30

20

10

0
0 500 1000 1500 2000 2500
Geometric mean fluorescence
of undivided cells

Figure 4.9.4 The relationship between the geometric mean fluorescence of CFSE in undivided
human PBMC and the response of CFSE-labeled PBMC to 5 μM phytohemagglutinin, measured
as the percentage of cells that have entered division after 4 days culture. Data were calculated as
described in Figure 4.9.2.

and the response of the CFSE-labeled cells to receptors that stimulate division in secondary
stimulation with 5 μM phytohemagglutinin. culture (Warren, 1999; Warren and Kinnear,
These data are a compilation of that obtained 1999).
in an experiment comparing labeling at differ-
ent CFSE concentrations (1 μM to 7.5 μM) Flow cytometry
and lymphocyte concentrations (0.5 × 106 to Analysis of in vivo–transferred cells. When
50 × 106 /ml), and in the presence or ab- examining CFSE-labeled lymphocytes in cell
sence of added protein. For any given CFSE suspensions from in vivo experiments, ideally
concentration, the presence of protein reduces 0.5 to 1 × 106 events are collected to ensure
the amount of CFSE incorporated. Labeling sufficient numbers of transferred cells are col-
is proportional to CFSE concentration and is lected for analysis (these can be ∼1% of the
essentially complete by 5 min. total population). Care should also be taken
Uniform labeling with CFSE is essential not to gate out the highly fluorescent CFSE-
to obtain clearly defined peaks following di- labeled cells during data acquisition. An alter-
vision. That is, the coefficient of variation native approach, which reduces data-storage
(CV) of the CFSE-labeled control unstimu- requirements, is to initially collect 25,000
lated lymphocytes must be small. This is in events in order to calculate the percentage of
part determined by the uniformity in cell vol- donor CFSE+ cells in the sample. One should
ume of the population, and in part by the tech- then collect 5000 CFSE+ events separately, us-
nique for mixing CFSE with the cells. For ing electronic gating, to allow an assessment
some applications it is useful to sort the cells of the division status and phenotype of CFSE-
for uniform staining following CFSE-labeling labeled cells.
(Nordon et al., 1999) prior to culture. Cul- In this way, CFSE can be used to easily
tured lymphocytes can also be labeled with identify the transferred cells, provided they
Use of CFSE to CFSE, but it is essential to label them when have not undergone extensive division. If,
Monitor
Lymphocyte they have reverted to small cells and are qui- on the other hand, CFSE is used to deter-
Migration and escent. CFSE-labeling of cultured human NK mine the proliferative response of the trans-
Proliferation cells has allowed an analysis of cell surface ferred cells, an independent marker, such as a
4.9.10
Supplement 84 Current Protocols in Immunology
CD45 allotype, should also be employed to al- to collect 5,000 to 10,000 events. For cultures
low definitive identification of the adoptively where cells have divided it is a good idea to col-
transferred cells from host cells, since CFSE lect as many as possible, e.g., 25,000 events.
fluorescence may be similar to background Comparative studies of CFSE-labeled lym-
auto-fluorescence in extensively divided cells. phocytes maintained in vitro or in vivo have
Figure 4.9.1 provides an example of how pro- shown that the labeled cells have identi-
gressive gating strategies can be used to dis- cal fluorescence-intensity characteristics; i.e,
criminate extensively divided CFSE-labeled CFSE-labeled lymphocytes lose the CFSE la-
cells from host cells. With in vivo experiments, bel at the same rate whether maintained in vitro
the number of events that need to be collected or in vivo (Lyons, 1999).
for analysis depends on four key factors: the Fluorescence compensation. Due to the po-
number of CFSE-labeled cells that have been tentially high fluorescence intensity of CFSE-
injected, the time since cell transfer, the organ labeled cells, significant “bleed over” can oc-
being examined, and the extent to which the cur in those detectors that detect emission
cells have divided. The latter factor is mainly wavelengths close to that of the CFSE/FITC
important if assessing cell division (not mi- channel. This is particularly notable in the de-
gration) is of prime importance, as each divi- tectors for PE (or propidium iodide). As such,
sion will result in a cell population of different it is important to have appropriate unstained
fluorescent intensity. Thus, to adequately re- and single-stained cell controls to apply appro-
solve each fluorescent peak, more events will priate pre- or post-acquisition compensation. It
need to be collected as more divisions (within should also be noted that at CFSE-labeling lev-
the detectable range) occur. Usually, the high- els required for detection of maximum cell di-
est proportion of CFSE-labeled lymphocytes visions, compensating out CFSE fluorescence
is detected in the peripheral blood and spleen, will be extremely difficult within the first 24
with a relatively low percentage of cells being hr after labeling and is not recommended. If
CFSE+ in the Peyer’s patches and peripheral examination of the CFSE-labeled cells is re-
lymph nodes. These considerations will influ- quired during this period, the lymphocytes
ence the number of events that need to be col- should be labeled with less CFSE [i.e., one-
lected to accurately determine the percentage quarter (1.25 μM) or one-eighth (0.625 μM)
of cells that are CFSE+ . Gating on CFSE+ the normal CFSE-labeling concentration].
cells and the collection of 5000 events per
fluorescent peak is a convenient means of ac-
quiring information about the CFSE+ lympho-
Anticipated Results
During the first few days following CFSE
cytes in cell suspensions, particularly when
labeling there is rapid decline in CFSE fluo-
there is a low frequency of CFSE+ cells.
rescence, presumably due to the loss of many
Analysis of in vitro–cultured cells. If re-
CF conjugates from the cells. Despite this loss
quired, add a known number of fluorescent
in fluorescence, there are sufficient amounts
counting beads to the sample prior to collec-
of long-lived conjugates remaining to allow
tion on the flow cytometer to allow subse-
nondividing CFSE-labeled lymphocytes to be
quent enumeration of cells in individual CFSE
tracked in vivo for up to 6 months following
peaks. Suitable beads are CaliBRITE beads
injection. If lymphocyte proliferation is to be
(Becton-Dickinson) and Flow-Count fluoro-
monitored, up to eight divisions can be dis-
spheres (Beckman Coulter). When analyzing
cerned for at least a week after cell transfer or
the data, apply appropriate electronic gates to
culture initiation. The slow decline in fluores-
view individually the live cells, apoptotic cells,
cence makes it increasingly difficult to mea-
and counting beads. The ratio of bead number
sure large numbers of cell divisions in ensuing
to cell number gives the actual cell concentra-
weeks.
tion in the sample.
For acquisition on the flow cytometer, the
logarithmic mode for the side-scatter (granu- Time Considerations
larity) parameter must be used with counting For a moderate-size experiment, the isola-
beads. Collect all events so that the dataset tion of lymphocytes, labeling, washing, and
includes beads, apoptotic cells, and live cells. setting up of lymphocyte cultures is accom-
Collect the samples at no more than 700 events plished in ∼4 hr. Harvesting and washing cells
per sec. Collect a sufficient number of events after culture takes ∼1 hr; an additional 2 to 3 hr
so that the proportion of cells in each peak can would be required for cell surface or intracel- In Vivo Assays
be accurately determined. For control cultures lular cytokine staining. Collecting cells on the for Lymphocyte
Function
where cells have not divided, it is adequate flow cytometer and data analysis take ∼4 hr.
4.9.11
Current Protocols in Immunology Supplement 84
Literature Cited ated anti-tumor immunity. J. Immunol. 16:6047-
Ford, W.L. 1978. The preparation and labeling 6055.
of lymphocytes. In Handbook of Experimental Nordon, R.E., Nakamura, M., Ramirez, C., and
Immunology, Vol. 3 (D.M. Weir, ed.) p. 23.1. Odell, R. 1999. Analysis of growth kinetics by
Blackwell Scientific, Oxford. division tracking. Immunol. Cell Biol. 77:523-
Garton, H.J. and Schoenwolf, G.C. 1996. Improv- 529.
ing the efficacy of fluorescent labeling for his- Oostendorp, R.A., Audet, J., and Eaves, C.J. 2000.
tological tracking of cells in early mammalian High-resolution tracking of cell division sug-
and avian embryos. Anat. Rec. 244:112-117. gests similar cell cycle kinetics of hematopoietic
Gett, A.V. and Hodgkin, P.D. 2000. A cellular stem cells stimulated in vitro and in vivo. Blood
calculus for signal integration by T cells. Nat. 95:855-862.
Immunol. 1:239-244. Parish, C.R. 1999. Fluorescent dyes for lymphocyte
Graziano, H.J., St-Pierre, Y., Beauchemin, C., migration and proliferation studies. Immunol.
Desrosiers, M., and Potworowski, E.F. 1998. Cell Biol. 77:499-508.
The fate of thymocytes labeled in vivo with Quah, B.J., Warren, H.S. and Parish, C.R. 2007.
CFSE. Exp. Cell Res. 240:75-85. Monitoring lymphocyte proliferation in vitro
Hasbold, J. and Hodgkin, P.D. 2000. Flow cytomet- and in vivo with the intracellular fluorescent dye
ric cell division tracking using nuclei. Cytometry carboxyfluorescein diacetate succinimidyl ester.
40:230-237. Nat. Protoc. 2:2049-2056.
Hasbold, J., Gett, A.V., Rush, J.S., Deenick, E., Stambas, J., Doherty, P.C., and Turner, S.J. 2007.
Avery, D., Jun, J., and Hodgkin, P.D. 1999. An in vivo cytotoxicity threshold for influenza
Quantitative analysis of lymphocyte differentia- A virus-specific effector and memory CD8(+)
tion and proliferation in vitro using carboxyflu- T cells. J. Immunol. 178:1285-1292.
orescein diacetate succinimidyl ester. Immunol. Sukkar, M.B., Stanley, A.J., Blake, A.E., Hodgkin,
Cell Biol. 77:516-522. P.D., Johnson, P.R., Armour, C.L., and Hughes,
Hawkins, E.D., Hommel, M., Turner, M.L., Battye, J.M. 2004. ‘Proliferative’ and ‘synthetic’ airway
F.L., Markham, J.F., and Hodgkin, P.D. 2007. smooth muscle cells are overlapping popula-
Measuring lymphocyte proliferation, survival tions. Immunol. Cell Biol. 8:471-478.
and differentiation using CFSE time-series data. Ueckert, J.E., Nebe von-Caron, G., Bos, A.P., and
Nat. Protoc. 2:2057-2067. ter Steeg, P.F. 1997. Flow cytometric analysis of
Hermans, I.F., Silk, J.D., Yang, J., Palmowski, M.J., Lactobacillus plantarum to monitor lag times,
Gileadi, U., McCarthy, C., Salio, M., Ronchese, cell division and injury. Lett. Appl. Microbiol.
F., and Cerundolo, V. 2004. The VITAL assay: 25:295-299.
A versatile fluorometric technique for assessing von Horsten, S., Helfritz, A., Kuhlmann, S.,
CTL- and NKT-mediated cytotoxicity against Nave, H., Tschernig, T., Pabst, R., Ben-
multiple targets in vitro and in vivo. J. Immunol. Eliyahu, S., Meyer, D., Schmidt, R.E., and
Methods 285:25-40. Schmitz, C. 2000. Stereological quantification
Jedema, I., van der Werff, N.M., Barge, R.M., of carboxyfluorescein-labeled rat lung metasta-
Willemze, R., and Falkenburg, J.H. 2004. New sis: A new method for the assessment of natural
CFSE-based assay to determine susceptibility to killer cell activity and tumor adhesion in vivo
lysis by cytotoxic T cells of leukemic precursor and in situ. J. Immunol. Methods 239:25-34.
cells within a heterogeneous target cell popula- Warren, H.S. 1999. Using carboxyfluorescein diac-
tion. Blood 103:2677-2682. etate succinimidyl ester to monitor human NK
Khil, L.Y., Kim, J.Y., Yoon, J.B., Kim, J.M., Keum, cell division: Analysis of the effect of activating
W.K., Kim, S.T., Yoon, Y., Yoon, M.Y., Moon, and inhibitory class I MHC receptors. Immunol.
C.K., Lee, J.H., Ha, J., Kim, S.S., and Kang, Cell Biol. 77:544-551.
I. 1997. Insulin has a limited effect on the cell Warren, H.S. and Kinnear, B.F. 1999. Quantitative
cycle progression in 3T3 L1 fibroblasts. Mol. analysis of the effect of CD16 ligation on hu-
Cell 7:742-748. man NK cell proliferation. J. Immunol. 162:735-
Lyons, A.B. 1999. Divided we stand: Tracking cell 742.
proliferation with carboxyfluorescein diacetate Weston, S.A. and Parish, C.R. 1990. New fluo-
succinimidyl ester. Immunol. Cell Biol. 77:509- rescent dyes for lymphocyte migration studies:
515. Analysis by flow cytometry and fluorescence
Lyons, A.B. 2000. Analyzing cell division in vivo microscopy. J. Immunol. Methods 133:87-97.
and in vitro using flow cytometric measurement
of CFSE dye dilution. J. Immunol. Methods Key References
243:147-154. Gett and Hodgkin, 2000. See above.
Lyons, A.B. and Parish, C.R. 1994. Determina- This paper is an elegant demonstration of the power
tion of lymphocyte division by flow cytometry. of CFSE to monitor lymphocyte division using a
J. Immunol. Methods 171:131-137. mathematical model for assessing the response of
Use of CFSE to
Monitor lymphocytes to different signals.
Marzo, A.L., Kinnear, B.F., Lake, R.A., Frelinger,
Lymphocyte J.J., Collins, E.J., Robinson, B.W., and Scott, Lyons and Parish, 1994. See above.
Migration and B. 2000. Tumor-specific CD4+ T cells have This is the original paper describing the use of
Proliferation a major “post-licensing” role in CTL medi- CFSE to monitor lymphocyte division.
4.9.12
Supplement 84 Current Protocols in Immunology
Parish, 1999. See above.
This paper reviews a range of fluorescent dyes for
their use in lymphocyte migration and analysis of
cell division.
Hawkins et al., 2007. See above.
An overview of the different approaches to analyze
CFSE data.

Internet Resources
http://www.vsh.com/
ModFit LT software is an excellent software for
cell proliferation analysis and is available for
Macintosh and PC.
http://www.flowjo.com/
FACS data analysis software, which is currently
available for Macintosh and PC.

In Vivo Assays
for Lymphocyte
Function

4.9.13
Current Protocols in Immunology Supplement 84

You might also like