Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Assessment of Phagocyte Functions by Flow UNIT 9.

19
Cytometry
Phagocytes are crucial to effective defense against infections. Neutrophilic polymor-
phonuclear leukocytes (PMNs), and monocytes and macrophages (MΦs), are crucial for
cellular defense against infections. Phagocyte functions encompass phagocytosis (attach-
ment and internalization), intra- and extracellular digestion of targets, oxidative burst, and
chemotaxis. Accurate measurement of phagocytosis in one cell population requires
discrimination from other populations by light scatter or fluorescent staining. Phagocy-
tosis of most microorganisms requires serum factors (opsonins). The major opsonins are
antibodies, complement factors, and fibronectin. Flow cytometry is applied to both routine
and experimental quantification of phagocyte functions as well as to the quantitation of
opsonin and antigen concentrations and activities. Bacteria, zymosan particles, and
microspheres are used as targets for different experimental and clinical purposes. The
basic protocols in this unit outline a four-step tandem procedure for evaluating all the
above-mentioned phagocyte functions: attachment and internalization (see Basic Protocol
1), processing of bacteria and zymosan particles (see Basic Protocol 2), phagocytosis and
oxidative burst (see Basic Protocol 3), and chemotaxis (see Basic Protocol 4).

PHAGOCYTOSIS (ATTACHMENT AND INTERNALIZATION) AND BASIC


PHAGOSOMAL pH PROTOCOL 1
This protocol is a two-step procedure for measurement of total phagocytosis (attachment
and internalization) and phagosomal pH. FITC-stained bacteria and zymosan particles
are used as targets, which are mixed with opsonins and leukocytes under controlled mixing
and temperature conditions. The first step concerns measurements of percentage phago-
cytosis, phagocyte fluorescence, and the loss of targets from the suspension, followed by
calculation of the phagocytic index.
In the second step, trypan blue is added after phagocytosis, and percentage phagocytosis
and phagocyte fluorescence are recorded. The numbers of attached and internalized
targets are calculated from the phagocytic index, the fluorescence of the phagocytes
before and after trypan blue quenching, and the fluorescence of targets. The difference in
the fluorescence between the internalized and the free, extracellular targets is taken as an
indirect measure of phagosomal pH using a standard curve of pH versus FITC fluores-
cence. The intensity of the phagocyte FITC fluorescence depends on complex intracellular
mechanisms that have to be taken into account when evaluating phagosomal pH (see
Commentary).
Materials
Targets:
Bacteria S. aureus Cowan III (NCTC 8532; see Support Protocol 4)
Zymosan particles (see Support Protocol 3 and/or 5)
Polychromatic red fluorescence (PC red)-dyed microspheres, 1-µm diameter
(Fluoresbrite Plain Microspheres, Polysciences) coated with outer membrane
vesicles from meningococci (OMV-beads; see Support Protocol 7)
Sørnes’s buffer (see recipe)
Opsonins (see Support Protocol 9)
Dihydrorhodamine 123 (DHR; see recipe)
White blood cell suspension (see Support Protocol 1)
Dulbecco’s phosphate-buffered saline (DPBS; see recipe) containing 0.02% (w/v)
EDTA, ice cold Studies of Cell
Function
Contributed by Carl-Fredrik Bassøe 9.19.1
Current Protocols in Cytometry (2002) 9.19.1-9.19.22
Copyright © 2002 by John Wiley & Sons, Inc. Supplement 21
Trypan blue
Vindeløv’s high-salt solution (see recipe)
Fluorescent beads (e.g., DNA-Check, Coulter)
Flow cytometer with 488-nm excitation and filters for the detection of green
fluorescence (R123, 505 to 545 nm), orange-red fluorescence (PC red beads,
560 to 590 nm), and red fluorescence (CD14-PE-Cy5, 660 to 700 nm)
Non-pyrogenic 96-well microtiter plates, sterile
12 × 75–mm tubes suitable for use on the flow cytometer

Prepare targets
1. Thaw selected targets. Calculate target concentration by flow cytometry using a
known target volume (VT), and a known volume (VL) and concentration (CL) of
leukocytes as internal reference, and note percent leukocytes (PL) and percent targets
(PT).
Target concentration = (PT/PL) × (CLVL/VT).
Targets are prepared and kept for several years. See specific support protocols for target
preparation.
2. Adjust bacteria and zymosan targets to 2.5 × 108 and 1.25 × 108 per milliliter in
Sørnes’s buffer, respectively.
Targets are adjusted to a useful concentration relative to the amount of added FITC, and
the applied quenching. The initial target/phagocyte (PMN + MΦ) ratio (R) is 20:1 for
bacteria and 10:1 for zymosan particles.

Incubate cells
3. Add 20 µl each of targets, diluted opsonins, and DHR working solution to each well
of 96-well microtiter plates, and adjust the final incubation mixture to 80 µl per well
by adding 20 µl Sørnes’s buffer. Use an additional duplicate plate (termed QUICK-
MIX) for controls. Add all the mentioned ingredients, except diluted opsonins, to the
QUICK-MIX plate.
4. Incubate 7.5 min at 37°C.
5. Add 20 µl white blood cell suspension to each well.
6. Incubate suspensions 7.5 min for phagocytosis, and 30 min for processing (see Basic
Protocol 2) under controlled mixing at 37°C.
7. Stop mixing, and immediately add 200 µl ice-cold DPBS containing 0.02% EDTA
to each well using the same sequence of additions as for the cells in order to equalize
phagocytosis time for all the wells. Add 20 µl diluted opsonins to the QUICK-MIX
plate.
8. Label three 12 × 75–mm tubes for each well. Add 400 µl DPBS with 0.02% EDTA
to one set, 400 µl of 2 mg/ml trypan blue in DPBS with 0.02% EDTA to the second
set, and 400 µl Vindeløv’s high salt-solution to the third set.
The first set of tubes is used for determination of percentage phagocytosis, phagocyte
fluorescence, target loss, and phagocytic index. The second set of tubes is used for
discrimination between attachment and internalization. The third set of tubes is reserved
for processing in Basic Protocol 2.
9. Mix the contents of each well in the 96-well plate thoroughly using a pipet (cells and
Assessment of targets sediment at different rates). For each well, transfer 100 µl suspension to each
Phagocyte of the three labeled tubes (step 8) corresponding to that well. Keep the tubes on ice
Functions by
Flow Cytometry and in the dark until flow cytometry measurement.
9.19.2
Supplement 21 Current Protocols in Cytometry
A
1023
L

Forward scatter

0
0 1023
Side scatter

B
1023 N1 N2
Forward scatter

N3 N4

0
100 101 102 103
FITC fluorescence

C
1023 O1 O2
Forward scatter

O3 O4

0
100 101 102 103
FITC fluorescence

Figure 9.19.1 The monocytes (M) and neutrophils (L) in the FS versus SS cytogram of leukocytes
phagocytosing bacteria or zymosan particles are gated to make separate FS versus FITC
fluorescence cytograms of monocytes and neutrophils (A). Percentage statistics and mean FITC
fluorescence values are recorded in the latter cytograms (B). The third cytogram (C) of FS versus
FITC fluorescence gives the relative counts of phagocytes and free targets. The phagocytic index
is calculated from the latter relative counts. Studies of Cell
Function

9.19.3
Current Protocols in Cytometry Supplement 21
10. Mix the contents of each tube thoroughly (cells and targets sediment at different rates)
just prior to running on the flow cytometer.

Acquire data
11. Calibrate flow cytometer with fluorescent beads (e.g., DNA-Check, Coulter). Use
electronic color compensation (UNIT 1.14) to minimize the spectral emission overlaps
between fluorochromes.
Coincidence rate of targets and leukocytes in the flow cell should be 1% to 2% (<5%).
12. Run the first set of tubes (DPBS with 0.02% EDTA only). Collect forward scatter
(FS), side scatter (SS), and yellow-green FITC fluorescence (FITC; Fig. 9.19.1).
Create ungated FS versus FITC cytogram (total cytogram).
13. Set region about noise. For the phagocytosis cytogram, make gated FS versus FITC
from cytogram by subtracting the noise region to create phagocytosis cytogram.
14. Create quadrant statistics in phagocytosis cytogram. Place the vertical limit close to
the left of the targets. Place the horizontal close beneath the lymphocytes. Record
quadrant statistics: region 1 contains non-phagocytes (N), region 2 contains phago-
cytes (F), and region 4 contains targets (T).
15. Record percentage phagocytosis:
P% = 100 × F%/(F% + N%).
16. Calculate average phagocytic index:
IP = R – (T%/F%).
R is the initial target/phagocyte (PMN + MΦ) ratio.
17. Note the mean fluorescence of the free, extracellular targets (FTe).

Create PMN and MΦ cytograms


18. Make gated FS versus FITC cytograms by gating on PMN and MΦ, respectively, in
the FS versus SS cytogram. Create quadrant statistics by placing the vertical and
horizontal limits as described in step 14.
19. Record quadrant statistics: region 1 contains non-phagocytosing PMNs or MΦs (N);
region 2 contains phagocytosing PMNs or MΦs (F).
20. Calculate PMN percentage phagocytosis:
PCPMN% = 100 × FCPMN%/(FCPMN% + NCPMN%).
21. Calculate MΦ percentage phagocytosis:
PCMΦ% = 100 × FCMΦ%/(FCMΦ% + NCMΦ%).
22. Record the mean fluorescence of the phagocytosing PMNs (FCPMN) and the mean
fluorescence of the phagocytosing MΦs (FCMΦ).

Determine attachment and internalization


23. Run the second set of tubes (trypan blue–containing samples) with the same flow
cytometer settings and record PMN percentage phagocytosis (PQPMN%).
24. Calculate percentage of PMNs with only adherent targets:

Assessment of PAPMN% = PCPMN% – PQPMN%.


Phagocyte 25. Record MΦ percentage phagocytosis (PQMΦ%).
Functions by
Flow Cytometry

9.19.4
Supplement 21 Current Protocols in Cytometry
26. Calculate percentage of MΦs with only adherent targets:
PAMΦ% = PCMΦ% – PQMΦ%.
27. Record the mean fluorescence of the phagocytosing PMN (FQPMN) and the mean
fluorescence of the phagocytosing MΦs (FQMΦ).
28. Calculate the mean number of targets attached to each PMN:
IAPMN = (FCPMN – FQPMN)/FTe.
29. Calculate the mean number of targets attached to each MΦ:
IAMΦ = (FCMΦ – FQMΦ)/FTe.
30. Calculate the mean fluorescence of the targets internalized by the PMNs:
FTiPMN = FQPMN/(IPMN × FTe).
31. Read the phagosomal pH off the pH versus fluorescence standard curve. Establish a
standard curve by measuring target fluorescence in suspensions with pH 4 to 10 at
0.5-pH step intervals.

PROCESSING OF BACTERIA AND ZYMOSAN PARTICLES BASIC


PROTOCOL 2
Phagocytes are added to Vindeløv’s high-salt solution containing the detergent NP40. In
a single step, phagocytes are lysed, most of the targets are liberated, and some adhere to

1000
digestion

100 Nc Nc − Z
EB fluorescence

10

.1
.1 1 10 100 1000
FITC fluorescence

Figure 9.19.2 Cytogram for the measurement of digestion. Cytogram of phagocytosing leuko-
cytes after the dissolution of the leukocyte cytoplasm in Vindeløv’s solution. Region Nc (EB-fluores-
cent nuclei) contains mainly lymphocyte nuclei. Region Nc + Z (EB- and FITC-fluorescent nuclei)
contains monocyte and neutrophil nuclei with 1 to 3 attached targets. Region Z contains free targets
(bacteria or zymosan particles). The mean EB and FITC fluorescence values are recorded from
Studies of Cell
region Z. Function

9.19.5
Current Protocols in Cytometry Supplement 21
the cell nuclei. Target DNA is stained with ethidium bromide (EB), and RNA is dissolved
by RNase. Flow cytometry measurements on FS, SS, and FITC and EB fluorescence can
be made on individual leukocyte nuclei and leukocyte nuclei with a few adherent targets,
as well as on the liberated, free targets and those that remained extracellularly during
phagocytosis.

Materials
Cells in Vindeløv’s high-salt solution from Basic Protocol 1, step 9
Flow cytometer with 488-nm excitation and filter set for detection of green (FITC)
and red (EB) fluorescence

A
103
CD14-PE-Cy5 fluorescence

102

101

A
100

0 1023
Side scatter

B
103
D1D2

102
R123 fluorescence

101

100

D3D4
100 101 102 103
PE-labeled OMV-beads

Figure 9.19.3 After the phagocytosis of OMV-beads, monocytes and neutrophils cannot be
discriminated by FS versus SS cytogram. Therefore, monocytes and neutrophils are stained with
PE-Cy5-labeled antiCD14 monoclonal antibody, and discriminated by PE-Cy5 fluorescence and
SS (A). Monocytes are positive for CD14 and are in region B, whereas, neutrophils are negative or
weakly CD14-positive, and located in region A. Monocytes and neutrophils are gated to separate
Assessment of cytograms (B) for measurement of phagocytosis (PE-fluorescence of OMV-beads) and oxidative
Phagocyte
Functions by burst (R123 fluorescence). The mean PE and R123 fluorescence of double-positive cells are
Flow Cytometry recorded from region D2, and that of single-positive cells from regions D1 and D4.

9.19.6
Supplement 21 Current Protocols in Cytometry
Acquire data

For bacteria targets


1a. Collect yellow-green FITC fluorescence and red EB fluorescence, and make log FITC
versus log EB cytogram. Place electronic gating region about individual cell nuclei
(Fig. 9.19.2, region Nc), cell nuclei with adherent targets (Fig. 9.19.2, region Nc +
Z), and free and liberated target cell nuclei (Fig. 9.19.2, region Z).
2a. Record mean fluorescence values on region Z in Figure 9.19.2.
3a. Compare mean green FITC and red EB fluorescence of QUICK-MIX with that of
PHS and PHS-IN.

For zymosan particles


1b. Make appropriate regions and collect information as in steps 1a to 3a (new settings
are required due to the higher FITC and EB fluorescence intensities of the zymosan
particles).
2b. Gate on region Z in Figure 9.19.2 into FS versus SS cytogram.
3b. Record mean FS and SS from the targets, and compare mean FS and SS of QUICK-
MIX (see below) with that of HYP and HYP-IN.

PHAGOCYTOSIS AND OXIDATIVE BURST BASIC


PROTOCOL 3
This protocol is used for the concurrent measurement of phagocytosis and oxidative burst.
Various microspheres can be used as targets (see Commentary). In this case, phagocytosis
is monitored by polychromatic red fluorescent (PC red) microspheres coated with outer
membrane vesicles from meningococci (OMV-beads). The oxidative burst is quantified
by the conversion of dihydrorhodamine123 (DHR) to green fluorescent rhodamine 123
(R123). PMNs and MΦs are discriminated using an orange fluorescent monoclonal
antiCD14 antibody. Data are simultaneously collected on targets, PMNs, and MΦs.
Additional Materials (also see Basic Protocol 1)
CD14-PE-Cy5 monoclonal antibody
Desired microspheres (see Support Protocol 7), adjusted to a concentration of 2.5
× 108 /ml in Sørnes’s buffer (see recipe); the initial microsphere/phagocyte
(PMN + MΦ) ratio (R) is 20:1.

Incubate cells
1. Follow Basic Protocol 1, steps 3 to 7.
2. Prepare microtiter plates in advance by adding 5 µl undiluted monoclonal antibody
to the desired number of wells. Add 100 µl cell suspension to each well, mix the
suspension gently, and incubate 1 hr in the dark on ice.
3. Mix the contents of each well thoroughly using a pipet and transfer to a 12 × 75–mm
tube containing 400 µl DPBS with 0.02% EDTA and proceed with flow cytometry.

Acquire data
4. Create cytogram of CD14 versus SS. Set separate regions about noise and small
particles, monocytes, and neutrophils. Also create a histogram on red fluorescence
(targets and phagocytes) gated from debris and small particles. Collect statistics on
free targets, and record mean target red fluorescence.

Studies of Cell
Function

9.19.7
Current Protocols in Cytometry Supplement 21
5. Create two cytograms to monitor phagocytosis and oxidative burst by monocytes and
neutrophils by placing quadrant statistics as described in Basic Protocol 1 (Fig.
9.19.3). Record the percentages in all regions.
6. Percentage phagocytosis P% = percentage of counts in region 2 plus region 4.
Percentage oxidative burst PO% = percentage of counts in region 1 plus region 2.
Percentage of inactive cells = percentage from region 3. Percentage of non-phagocy-
tosing cells producing oxidative burst = percentage from region 1. Percentage of
phagocytosing cells without oxidative burst = percentage from region 4.
7. Record the mean green fluorescence in region 1 (oxidative burst alone), green and
red fluorescence in region 2 (oxidative burst and phagocytosis), and red fluorescence
in region 4 (phagocytosis only).
8. For region 2 in cytograms from both monocytes and neutrophils, calculate phagocytic
index IPMN (IMΦ) = mean red fluorescence of phagocytes in region 2 divided by the
red fluorescence of the free targets.
9. For region 2, calculate neutrophil oxidative ratio OPMN = mean green fluorescence of
phagocytes in region 2 divided by IPMN, and monocyte oxidative ratio OMΦ = mean
green fluorescence of phagocytes in region 2 divided by IMΦ.

BASIC CHEMOTAXIS
PROTOCOL 4
Measurement of chemotaxis is based on the selective, active, and stimulated motion of
neutrophils across a 3-µm filter. The source population serves as a reference, and
non-stimulated cells as a control. Two or more chemotactic stimuli, zymosan-activated
serum (ZAS) and formyl-methionyl-leucyl-phenylalanine (fMLP), are routinely used.
Materials
White blood cell suspension (see Support Protocol 1)
Sørnes’s buffer (see recipe)
DPBS (see recipe) containing 0.2% (w/v) EDTA
5% zymosan-activated serum (ZAS; see Support Protocol 3)
Paraformaldehyde (optional)
12 × 75–mm tubes
24-well transwell chemotaxis plates (6.5-mm diameter, 3-µm pore size; Corning)
37°C, 5% CO2 incubator

Prepare cells
1. Centrifuge white blood cell suspension 5 min at 350 × g, 20°C, and resuspend in
Sørnes’s buffer to a final concentration of 5 × 106 leukocytes/ml.
2. For reference sample, add 2 ml DPBS containing 0.2% EDTA, 100 µl cell suspension,
and 500 µl Sørnes’s buffer to each of two 12 × 75–mm tubes.
These tubes contain the same volume as the final dilution in the lower well, and are the
100% cell count.
3. Add 600 µl Sørnes’s buffer to the lower chamber of two wells of the 24-well transwell
microtiter plate (control).
4. Add 600 µl of 5% ZAS (or other stimulus) in Sørnes’s buffer to the lower chamber
of two wells of the 24-well transwell microtiter plate (control).
Assessment of 5. Add 100 µl cell suspension (5 × 106 leukocytes/ml) to the corresponding upper
Phagocyte chambers of the 24-well transwell microtiter plate.
Functions by
Flow Cytometry

9.19.8
Supplement 21 Current Protocols in Cytometry
6. Incubate 30 min in a 37°C, 5% CO2 incubator. After the incubation, immediately
wash upper and lower chambers each with 2 ml ice-cold DPBS containing 0.2%
EDTA, and transfer the leukocytes to 12 × 75–mm tubes. Keep the cell suspensions
on ice and proceed with flow cytometry, or fix cells in 1.0% final paraformaldehyde
for later investigation.

Acquire data
7. Record total and differential counts of lymphocytes, MΦs, and PMNs using FS
versus SS and appropriate gates from reference sample and the lower chamber.
8. Calculate spontaneous (random) motion = 100 × PMN count in lower chamber
without stimulus divided by PMN count in reference suspension.
9. Calculate chemotaxis = 100 × PMN count in lower chamber with stimulus divided
by PMN count in reference suspension.

PREPARATION OF WHITE BLOOD CELL SUSPENSION SUPPORT


PROTOCOL 1
This protocol describes the isolation of a white blood cell suspension from peripheral
blood. The suspension provides starting material for the basic protocols.

Materials
Lysing solution (see recipe)
Dulbecco’s phosphate buffered saline with glucose and BSA (DPBS-GA; see
recipe)
Vacutainer tubes containing 100 U preservative-free heparin (UNIT 9.7)
50-ml centrifuge tubes, sterile
Additional materials for blood collection
1. Collect peripheral blood sample in the Vacutainer tube containing heparin.
2. In a 50-ml centrifuge tube, dilute heparinized blood 1:10 (v/v) in lysing solution, mix
gently, and leave 10 min at room temperature.
3. Centrifuge 5 min at 350 × g, room temperature, resuspend the pellet in 20 ml washing
solution, and centrifuge again 5 min at 350 × g, room temperature.
4. If lysis is incomplete, resuspend the pellet 5 min in NH4Cl solution (final 8 mg/ml),
pH 7.4, centrifuge 5 min at 350 × g, room temperature, and resuspend in 20 ml
DPBS-GA.
5. Centrifuge 5 min at 350 × g, room temperature, and resuspend pellet in 1 ml
DPBS-GA per 10 ml of starting heparinized blood.
6. Adjust the concentration to 1.25 × 107 white cells/ml in DPBS-GA.

Studies of Cell
Function

9.19.9
Current Protocols in Cytometry Supplement 21
SUPPORT PREPARATION OF SERUM FROM PERIPHERAL BLOOD
PROTOCOL 2 Blood is collected in serum tubes with no anticoagulant and is allowed to stand undis-
turbed until it clots. The clot is removed and the remaining liquid is centrifuged to remove
any residual erythrocytes and debris. The resulting serum is divided into aliquots and
frozen.
Materials
Freshly extracted peripheral blood
Red-top collection tubes (no anticoagulant)
50-ml centrifuge tubes
Additional equipment for venipuncture
1. Place tube of blood in a rack and allow to sit undisturbed until a clot forms.
Blood must be collected without an anticoagulant.
If a clot does not start to form within 15 min, initiate clotting by inserting a wooden
applicator stick into the tube. Then begin with step 1.
2. Using a wooden applicator stick, gently loosen the clot and remove from the tube.
Do not break up clot.
3. Transfer serum to 50-ml centrifuge tube and centrifuge 10 min at 2700 × g, 4°C.
4. Save the supernatant.
5. Combine the supernatants from at least 7 different donors, divide into 500-µl aliquots
or other desired volume, and store in appropriate containers at –20°C.
Do not subject sera to repeated freeze-thaw cycles.

SUPPORT ZYMOSAN-ACTIVATED SERUM (ZAS) AND PREOPSONIZED ZYMOSAN


PROTOCOL 3 PARTICLES
ZAS is used as one stimulus for cell motility in the chemotaxis assay. Preopsonized
zymosan particles are used in the assay for complement-mediated phagocytosis. ZAS and
complement fragment C3b- and C3bi-opsonized zymosan particles are prepared simul-
taneously.
Materials
Zymosan A particles (Sigma)
DPBS (see recipe)
Fresh human serum (see Support Protocol 2)
Sørnes’s buffer (see recipe)
15-ml plastic centrifuge tube
37°C incubator with rotator
1. Add 100 mg zymosan A particles and then 10 ml DPBS to a 15-ml plastic centrifuge
tube and shake vigorously.
2. Centrifuge 10 min at 2000 × g, 20°C.
3. Pour off supernatant and add 12.5 ml fresh human serum. Shake and rotate solution
1 hr at 37°C.
4. Centrifuge 10 min at 2000 × g, 20°C, decant serum and label as ZAS, and store up
to 5 years at –80°C.
Assessment of 5. Resuspend zymosan A pellet in 10 ml Sørnes’s buffer. Use immediately or store as
Phagocyte
Functions by preopsonized zymosan particles in 1-ml aliquots up to 5 years at –80°C.
Flow Cytometry

9.19.10
Supplement 21 Current Protocols in Cytometry
FITC-LABELING OF BACTERIA SUPPORT
PROTOCOL 4
FITC-labeled bacteria are used to investigate adhesion of bacteria to leukocytes, and
antibody-dependent and antibody-plus-complement-stimulated phagocytosis.
Materials
Fluorescein isothiocyanate (FITC)
96% and 70% ethanol
Heart infusion broth (HIB; Difco)
S. aureus Cowan III (NCTC 8532 O/N)
Blood agar plates (e.g., Becton Dickinson)
0.9% (w/v) NaCl
Sørnes’s buffer (see recipe)
Spectrophotometer
1. Weigh out 10 mg FITC, and add to 0.1 ml of 96% ethanol. Dilute the FITC-ethanol
solution to 100 ml in heart infusion broth (HIB).
2. Grow S. aureus Cowan III on blood agar, and add bacteria to ∼2 ml HIB. Mix until
the suspension is free from clumps. Warm 100 ml HIB to 37°C and add the 2-ml S.
aureus suspension. Read the OD using a spectrophotometer (the OD of the suspension
should be 0.05 to 0.1 at 620 nm). Incubate at 37°C until OD = 1.0 (∼2.5 hr).
3. Pellet the bacteria by centrifuging 10 min at 2000 × g, room temperature. Resuspend
bacteria in ∼2 ml of 0.9% NaCl. Mix until there are no visible bacteria clumps.
4. Dilute bacteria in ∼90 ml ice-cold 70% ethanol to OD=1.0. Let the bacteria fix 30
min at 0°C. Repeat step 3 three times at 4°C.
5. Dilute the bacteria in two 50-ml tubes each containing 30 ml FITC solution (see step
1). Incubate 30 min at 37°C with continuous mixing.
6. Repeat step 3 three times. Resuspend bacteria in Sørnes’s buffer, and adjust to OD =
1. Divide suspensions into 0.5-ml aliquots and store FITC-stained bacteria up to 5
years at –80°C.

FITC-LABELING OF ZYMOSAN PARTICLES SUPPORT


PROTOCOL 5
Phagocytosis is mediated by complement fragments, mainly C3b and C3bi. Zymosan
particles bind complement factor C3 that is converted into C3b and C3bi, which remain
covalently bound on the particles’ surface. FITC-labeled zymosan particles are used to
measure complement-mediated phagocytosis, and intracellular processing mediated by
complement.
Additional Materials (also see Support Protocol 4)
Zymosan A particles
DPBS (see recipe)
10-ml glass tubes
0.2-µm filter
End-over-end rotator, 37°C
1. Add 100 mg zymosan A particles to each of ten 10-ml glass tubes.
2. Make FITC solution: Place 10 mg FITC in a 10-ml glass tube. Add 0.5 ml of 96%
ethanol to dissolve the FITC. Add 9.5 ml DPBS at 37°C to a final 1 mg/ml FITC
stock solution. Dilute with DPBS at 37°C to a working solution containing 0.05 Studies of Cell
mg/ml FITC. Function

9.19.11
Current Protocols in Cytometry Supplement 21
3. Filter sterilize through a 0.2-µm filter.
4. Add 10 ml of 0.05 mg/ml FITC solution to each glass tube with zymosan particles.
Incubate 30 min at 37°C with end-over-end rotation.
5. Centrifuge 10 min at 2000 × g, room temperature, and resuspend the pellet in DPBS.
Repeat three times.
6. Centrifuge 10 min at 2000 × g, 20°C, and resuspend the pellet in 200 ml Sørnes’s
buffer. Store 0.5-ml aliquots protected from light up to 5 years at –80°C.

SUPPORT DILUTIONS OF FITC-LABELED ZYMOSAN A PARTICLES


PROTOCOL 6
This protocol is used for the adjustment of FITC concentration, in case the FITC-stained
particles are not quenched by trypan blue.
Additional Materials (also see Support Protocol 5)
Trypan blue
15-ml polypropylene tubes
1. Add 100 mg zymosan A particles and 2 ml DPBS to a 15-ml polypropylene tube. Mix
with a pipet or ultrasound (30 sec) until the suspension is free from clumps. Dilute
to 10 ml with DPBS. Centrifuge 10 min at 2000 × g, room temperature, and discard
the supernatant.
2. Dissolve 0.25 mg FITC in 0.1 ml of 96% ethanol and immediately dilute in 1 ml
DPBS at 37°C. Wait until all FITC is solubilized. Pass through a 0.22-µm filter. Make
the following concentrations of FITC in DPBS: 0.25, 0.1, 0.05, and 0.025 mg/ml.
3. Resuspend zymosan A pellet in a little FITC solution and mix until free from clumps.
Thereafter, add 10 ml FITC solution per 100 mg zymosan particles. Incubate with
end-over-end rotation 30 min at 37°C.
4. Centrifuge 10 min at 2000 × g, room temperature. Decant and resuspend in 10 ml
room temperature 0.9% NaCl. Repeat this step three times.
5. Resuspend the pellet in 10 ml DBPS. Count zymosan particles using flow cytometry
(see Support Protocol 8) and dilute to 5 × 108 particles/ml.
6. Run suspension on flow cytometer and place the modal FITC fluorescence in channel
∼100.
7. Add 100 µl FITC-labeled zymosan suspension to 400 µl trypan blue suspension. Run
on flow cytometer. Use the solution with the highest FITC concentration that is 100%
effectively quenched.
8. Store the selected FITC-stained zymosan population in 1-ml aliquots up to 5 years
at –80°C.

Assessment of
Phagocyte
Functions by
Flow Cytometry

9.19.12
Supplement 21 Current Protocols in Cytometry
ANTIGEN COATING OF POLYSTYRENE BEADS SUPPORT
In principle, beads are incubated with an excess of antigen overnight at room temperature PROTOCOL 7
(20°C). Unreacted sites on the beads are blocked with 2% BSA. Antigen-coated beads
are suspended in a storage buffer, and kept protected from daylight at 4°C.
OMV from N. meningitidis strain 44/76 (B:15:P1.7,16) is prepared as for vaccine
production (Lehmann, 1997).
Materials
Fluoresbrite PC red 1.0-µm microspheres (Polysciences)
0.1 M borate buffer, pH 8.5 (0.1 M boric acid; Polysciences)
OMV-beads with and without 2% (w/v) bovine serum albumin, endotoxin-free
(BSA; Roche Diagnostics)
Sodium phosphate storage buffer (Polysciences)
End-over-end rotator
1. Centrifuge 500 µl Fluoresbrite PC red microspheres (4.55 × 1010 microspheres/ml)
5 min at 15,600 × g, room temperature. Resuspend pellet in 550 µl of 0.1 M borate
buffer, pH 8.5.
2. Add 450 µl of a 0.94 mg/ml (saturating) OMV suspension. Mix by end-over-end
rotation 20 hr in the dark at room temperature.
3. Centrifuge OMV-beads 5 min at 15,600 × g, room temperature, and resuspend them
in OMV suspension containing 2% endotoxin-free BSA. Incubate with end-over-end
rotation 20 hr at 20°C.
4. Centrifuge OMV-beads with blocked free surface sites 5 min at 15,600 × g, room
temperature, and resuspend in sodium phosphate storage buffer. Store up to 1 year at
4°C.

COUNTING TARGETS SUPPORT


For the quantification of small particles, leukocytes of a known concentration are used as PROTOCOL 8
reference. Mix known volumes of leukocytes and beads. Discriminate leukocytes, debris,
and fluorescent beads by forward and side scatter. Place electronic windows around
targets and leukocytes and record the relative count of beads and leukocytes. Calculate
the concentration of beads by multiplying the concentration of leukocytes by the relative
bead/leukocyte ratio.

OPSONINS SUPPORT
The phagocytosis of many pathogenic microorganisms and fungi requires serum opson- PROTOCOL 9
ins. Complement is a natural opsonin for many microorganisms and fungi. Complement
factor 3 (C3) binds to the surface of many non-capsulated microorganisms and zymosan
particles. C3 is converted to C3b and C3bi that remain attached to the targets, and bind
to complement receptors on phagocytes. For example, C3bi binds to complement receptor
3(CD11b). Hypo- and agammaglobulinemic sera are useful sources of complement. Due
to the low antibody concentration, they are used in conjunction with zymosan particles
to monitor complement-mediated phagocytosis.
Infections normally induce the synthesis of antibodies. In the acute phase (day 0 to 2),
the concentration of specific antibodies against the infectious agent is low (acute-phase
serum). After 2 to 4 weeks, the concentration of specific antibodies is high (convalescent
serum). Antibodies bind to the microorganisms by the Fab part, and link them to the
phagocytes through the Fc part. The Fc part of IgG promotes phagocytosis by binding to
Fc-receptors FcγRII (CD32) and FcγRIII (CD16) on neutrophils, and FcγRII (CD32) on Studies of Cell
Function

9.19.13
Current Protocols in Cytometry Supplement 21
monocytes. Convalescent serum is used as a source of specific antibodies. Acute-phase
serum serves as control.

Materials
Healthy human serum
Hypogammaglobulinemic serum
Acute-phase and convalescent serum
Sørnes’s buffer (see recipe)

For a pooled serum from healthy controls


1a. Collect serum (see Support Protocol 2) from healthy individuals, and ensure that CH50
is within the normal reference range.
2a. Mix equal volumes of serum from each of seven healthy individuals and label as
pooled human serum (PHS). Heat-inactivate a portion (step 3a) and store the
remainder in 0.5-ml aliquots up to 5 years at –80°C.
3a. Inactivate complement by heating 30 min at 56°C. Label as PHS-IN and immediately
store in 0.5-ml aliquots up to 5 years at –80°C.
4a. Thaw shortly before use.

For hypogammaglobulinemic serum


1b. Collect hypogammaglobulinemic serum (HYP) from agammaglobulinemic patients
before immunoglobulin substitution. Heat-inactivate a portion (step 2b) and store the
remainder in 0.5-ml aliquots up to 5 years at –80°C.
2b. Inactivate complement by heating 30 min at 56°C. Label as HYP-IN. Immediately
after preparation store in 0.5-ml aliquots up to 5 years at –80°C.
3b. Thaw shortly before use.

For acute-phase and convalescent serum


1c. For antigen- and antibody-dependent phagocytosis: collect acute-phase serum (APS)
from patient on admission, and convalescent serum (REC), 6 weeks later. Heat-inac-
tivate a portion (step 2c) and store the remainder in 0.5-ml aliquots at –80°C.
In the example presented here, sera collected at admission and 6 weeks later from a patient
with meningococcal C:15:P1.7,16 disease are used.
2c. Inactivate complement by heating 30 min at 56°C. Label as REC-IN. Immediately
after preparation store in 0.5-ml aliquots up to 5 years at –80°C.
3c. Thaw shortly before use.
4c. Dilute 0.5 ml of each serum (APS, REC) 1:4 in Sørnes’s buffer.
OMV is obtained from serogroup B meningococci vaccination strain 44/76, B:15:P1.7.16.
OMV-suspension consists of 50 ìg protein and 3.5 ìg lipopolysaccharide per ml suspen-
sion.

Assessment of
Phagocyte
Functions by
Flow Cytometry

9.19.14
Supplement 21 Current Protocols in Cytometry
REAGENTS AND SOLUTIONS
Use deionized, distilled water in all recipes and protocol steps. For common stock solutions, see
APPENDIX 2A; for suppliers, see SUPPLIERS APPENDIX.

Dihydrorhodamine (DHR) 123


Stock solution: 10 mg/ml in dimethylsulfoxide (DMSO). Store protected from light
≤3 weeks at –80°C.
Working solution: Dilute 1:1000 in Sørnes’s buffer (see recipe) before use.
Dulbecco’s phosphate buffered saline (DPBS)
Make 1 liter:
8 g NaCl
1.44 g Na2HPO4.2H2O
0.2 g KCl
0.2 g KH2PO4
Adjust to pH 7.4
Store ≤2 months at 4°C
Dulbecco’s phosphate buffered saline with glucose and BSA (DPBS-GA)
100 ml DPBS (see recipe)
0.5 g BSA
0.1 g glucose
Adjust to pH 7.4
Make fresh daily
Lysing solution
Make 1 liter:
8 g NH4Cl
0.8 g NaHCO3
0.88 g EDTA.2H2O
pH ≈ 6.8
Store ≤2 months at 4°C
Sørnes’s buffer
100 ml DPBS-GA (see recipe)
13.2 mg CaCl2.2H2O
12.1 mg MgSO4.7H2O
Adjust to pH 7.4
Make fresh daily
Vindeløv’s high-salt solution
Make 1 liter:
0.01 M glycine
0.7507 g NaOH
9.8575 mg ethidium bromide (2.5 × 10-5 M)
1 ml Nonidet P-40 (0.1%, v/v)
17.532 g NaCl (0.3 M)
pH should be 10
Store up to 1 year at 4°C.
Just before use, add 700 U/liter ribonuclease from 100 U/ml stock (see recipe).
Ribonuclease
Stock solution 1 mg/ml = 100 U/ml
Dissolve in 50 mM Tris⋅Cl, pH 8 (APPENDIX 2A). Store in 0.5-ml aliquots at –80°C.
Studies of Cell
Function

9.19.15
Current Protocols in Cytometry Supplement 21
COMMENTARY
Background Information criminated from cellular defects. If a deviation
from the expected values is observed, it is
General considerations impossible to know whether the unexpected
Many specific disorders of phagocyte func- results are caused by serum opsonins or leuko-
tions are known, such as chronic granuloma- cytes. In such instances, the present methods
tous disease, CD11b/CD18 deficiency, lazy are needed as controls. It should also be noted
leukocyte, and Chediak-Higashi syndrome. that diminished phagocyte function can be
These are hereditary syndromes that manifest compensated by effects of serum factors. Even
themselves in childhood. However, little is when the overall results are within the reference
known concerning phagocyte functions in range, whole-blood assays may miss important
common, acquired clinical disorders of adults deviations from the reference range.
and the elderly. Surprisingly little has been
done on leukemia, myelodysplasia, and bone Targets
marrow failure. For example, up to the year The purpose of the investigation determines
2000, only 14 investigations of phagocyte func- the choice of target. Since sera and targets vary,
tions in leukemia were available. Therefore, it there is no universal calibration method for
is difficult to advise on the selection of useful phagocytosis.
methods in acquired immunodeficiencies re-
lated to phagocyte functions. Zymosan particles
Several methods are available for the moni- In brief, zymosan particles are phagocyto-
toring of phagocyte functions. The present unit sed by complement, mainly C3bi, and are se-
aims at in-depth investigations of phagocyte lected to monitor serum C3 opsonic activity and
functions that reflect the tandem attack and complement receptor activity (mainly
processing of pathogenic microorganisms. Ba- CD11b/CD18). In HYP, 95% to 100% of PMNs
sic Protocols 1, 3, and 4 can be applied as and MΦs phagocytose zymosan particles. With
stand-alone procedures. Digestion cannot be HYP-IN, neither PMNs nor MΦs phagocytose
monitored without knowing that phagocytosis zymosan particles.
has taken place to a sufficient degree to monitor Two-color investigations using directly con-
intracellular processing. Therefore, if Basic jugated antibodies show that all the PMNs and
Protocol 2 is applied, Basic Protocol 1 must be MΦs from the same suspensions carry comple-
used in conjunction. ment receptor 1 (CR1, CD35) and complement
All four basic protocols can be used together receptor 3 (CR3, CD11b/CD18).
on a routine basis. The protocols are fine-tuned Both live and dead microorganisms can be
to minimize the workload. For example, 20 µl used. S. aureus is used to monitor serum opson-
of each reagent is added to all microwells to ins (mainly IgG), Fcγ-receptors, and the coop-
minimize preparation time, and the same buffer eration between IgG and complement. PHS and
(DPBS) is used for cell preparations, dye and PHS-IN are used for combined IgG-comple-
serum dilutions, and all incubations. ment phagocytosis and IgG-mediated phago-
All investigations are done in duplicate. Re- cytosis, respectively. Assessment of phagocy-
producibility is excellent. Day-to-day and in- tosis using microorganisms is influenced by
terindividual variations with leukocytes from properties other than antigenic properties of
healthy individuals are small. microorganisms. Intracellular sorting in MΦs,
e.g., differs between various strains of the same
Less laborious methods microbial species.
Simpler methods are available, but these rely Microorganisms containing green fluores-
on assumptions that require careful controls. cent protein (GFP) are widely used for moni-
Technical details that might seem trivial are toring phagocytosis. Some technical problems
quantitatively important. The most important have not been addressed. For example, the kill-
caveats are addressed below. ing and digestion of GFP by powerful phago-
Time-consuming isolation of leukocytes has cyte enzymes is usually not taken into account.
been a major concern in working with flow Caution is required with interpretation of the
cytometry and phagocytosis. Whole-blood results of fluorescence measurements.
Assessment of methods have the advantage of being the least
Phagocyte time consuming, and are useful in screening.
Functions by The effect of serum opsonins cannot be dis-
Flow Cytometry

9.19.16
Supplement 21 Current Protocols in Cytometry
Microspheres Opsonins
Coated beads have been used in phagocy- There is ample proof that phagocytosis of
tosis assays for a long time, but the use of zymosan particles, many strains of S. aureus,
bead-associated antigens in the quantification N. meningitidis, and many other bacteria spe-
of phagocytosis is of recent date (Bassøe et al., cies does not occur in the absence of opsonins.
2000). The advantage of using fluorescent The importance of attaching opsonins to suit-
beads lies in the ability to quantify the phago- able antigens is underestimated. Passive ad-
cytic and oxidative responses in mixed cell sorption of opsonins to microspheres results in
populations. feeble phagocytosis. Target/phagocyte ratios
Microspheres can be seen as surrogate mi- >50:1 and incubation times >30 min are re-
croorganisms, and are useful for the study of quired to observe phagocytosis. Under these
specific opsonins and antigens. Various types conditions, percentage phagocytosis is only
of coated beads have been used in phagocytosis ≤15%, and the phagocytic index ∼1 to 3. This
assays (reviewed in Bassøe et al., 2000): latex- low rate of phagocytosis is probably due to the
IgG, latex-IgG-C3, microspheres-C3b, micro- attachment of the Fcγ-part of the IgG molecules
spheres-iC3b, beads bearing ligands for to the microsphere surface, making them un-
CD11b, latex-lysozyme, and OMV-beads. available to Fcγ receptors on the leukocyte
Any of these coated beads might be used in surface.
combined assays for monocyte and neutrophil
functions. OMV-beads mimic N. meningitidis. Antigens
Phagocytosis and oxidative burst mediated by Opsonins bind to antigens attached to the
OMV-beads require the presence of specific surface of targets. C3bi and the Fcγ-fragment
antimeningococcal antibodies and the appro- of IgG extend into the medium, making them
priate antigen. REC and APS are used for com- easily available for complement receptors and
bined IgG-complement phagocytosis and IgG- Fcγ-R, respectively.
mediated phagocytosis, respectively. The same
principle can be applied to other antigens that Phagocytic index
can be attached to polystyrene microspheres. Flow cytometric measurements of the
phagocytic index have adapted the principles
Attachment and internalization introduced by Leishman (1927) and Maaløe
Various agents have been used to discrimi- (1946). Leishman’s method is based on count-
nate attached from internalized particles. The ing the number of targets associated with each
enzyme lysostaphin, which dissolves free and cell. This is equivalent to measuring phagocyte
attached staphylococci, does not penetrate neu- fluorescence along the fluorescence dimension
trophils, and does not digest internalized bac- of the correspondingly labeled targets.
teria. Crystal violet is used to quench attached Maaløe’s method depends on counting the
targets, but diffuses into phagocytes and number of free targets before and after phago-
quenches intracellular targets too. For this rea- cytosis. Leishman’s and Maaløe’s methods
son, crystal violet should be abandoned and have advantages and disadvantages.
replaced with trypan blue, which does not enter (1) Leishman’s approach is applicable to
phagocytes. fluorescent microspheres. The stability of the
fluorescence is due to incorporation of dye in
Attachment the solid polymer. The fluorescence intensity is
The percentage of phagocytosing PMNs and unaffected by enzymes, toxic radicals, and pH.
MΦs is of the same order of magnitude before Thus, the mean phagocytic index can be easily
and after trypan blue quenching. Thus, PMNs and accurately calculated by dividing the mean
and MΦs do not carry targets only on the fluorescence of each phagocyte subpopulation
surface. by the mean fluorescence of the free micro-
As a rule, 0.5 to 2.0 targets are attached to spheres. When only one subpopulation of
the outer side of the plasma membrane. phagocytes is present, the phagocytic index
calculated from microsphere fluorescence
Internalization closely correlates with the index obtained from
The remaining targets associated with the loss of particles from the suspension
PMNs or MΦs are internalized. These results (Lehmann et al., 1997).
are obtained whether phagocytosis is sustained (2) Maaløe’s approach overcomes the prob-
by PHS or complement is inactivated (PHS- lem that the phagocytic index cannot be quan- Studies of Cell
IN). tified by the FITC fluorescence of the phago- Function

9.19.17
Current Protocols in Cytometry Supplement 21
cytes containing FITC-labeled targets (Bassøe Phagosomal pH
et al., 1983). Although that approach adds some Phagocyte FITC fluorescence depends on
technical inconveniences (see Troubleshoot- quenching, digestion and release of florescent
ing), it is mandatory for accurate measure- molecules (amino acids, peptides, proteins, and
ments. glucosaminoglycans) from stained targets,
electrolyte composition and concentrations,
Critical Parameters and pH.
Clearly, the estimation of phagosomal pH
Fluorescence measurements using FITC fluorescence should be interpreted
Phagocyte FITC fluorescence has been used with care. However, little digestion of phago-
as a measure of phagocytosis. This is a simple cytosed material takes place during the first 15
approach that gives an overall impression of the min of incubation (Bassøe, 1984). At 15 min,
combined fluorescence of attached and inter- the modal FITC fluorescences of liberated and
nalized targets. The phagosomal environment free FITC-labeled particles are similar (Bassøe,
profoundly affects target fluorescence. Some 1984). At 7.5 min, the fluorescence intensity of
fluorescence changes persist after the targets liberated, FITC-stained targets is similar to that
are liberated from the phagocytes (Bassøe, of the free FITC-labeled particles. Thus, within
1984; Hurst et al., 1984). In order to overcome this short incubation time, the changes in FITC
the influence of phagosomal pH on fluorescent fluorescence of phagocytosed targets are re-
labels like FITC, phagocytes have been fixed versible. FITC fluorescence is a sensitive pH
and permeabilized (Cantinieaux et al., 1989). indicator (Bassøe, 1984). Accordingly, with
However, FITC molecules may be chemically incubation times <15 min, total phagocyte fluo-
modified by the oxidative burst and other intra- rescence can be used as a (semiquantitative)
cellular chemical reactions (Hurst et al., 1984). measure of phagosomal pH.
Fluorescent molecules attached to digestible
targets may be released from the targets by Mixing
phagosomal enzymes (Bassøe, 1984). Perme- The frequency of hits between targets and
abilization may liberate digested, small fluo- phagocytes influences the rate of phagocytosis,
rescent amino acids, peptides, and glycosami- and the phagocytosis index is profoundly af-
noglycans from the cells. However, digestion fected by mixing (Bassøe, 2000). Several mix-
can barely be measured with <15-min incuba- ing procedures have been used: periodical stir-
tion, and only a few fluorescent molecules are ring, end-over-end rotation, shaking, and auto-
expected to be released with shorter incubation mated microtiter plate mixers.
times. Periodical stirring and shaking are unde-
Fluorescence measurements may underesti- fined terms and should be avoided. End-over-
mate phagocytic indices. In general, the esti- end rotation is used in flow cytometry phago-
mation of the phagocytic index from the fluo- cytosis assays to secure optimal mixing. The
rescence of digestible targets or pH-sensitive disadvantage is the large volume and high cell
fluorescent probes is discouraged. numbers (∼5 × 106 phagocytes) required. In
order to miniaturize phagocyte assays, and at
QUICK-MIX controls the same time standardize mixing, use 96-well
The fluorescence of FITC molecules is in- microwell plates and standard mixing.
fluenced by serum factors other than electro- Considering its importance, mixing of tar-
lytes. Proteins, for example, may diminish the gets and leukocytes is taken on too easily. Vast
fluorescence intensity of FITC. The controls differences are reported in assays with corre-
overcoming these problems are nicknamed sponding concentrations and activities of tar-
QUICK-MIX. QUICK-MIX controls are not gets, phagocytes, and opsonins, but differences
subject to phagocytosis, but all the other incu- in mixing (see Bassøe, 2000). Some commer-
bation steps are followed. There is only one cially available mixers are easy to use, the rate
exception. QUICK-MIX is not required for of mixing can be controlled, and temperature
trypan blue–quenched samples of zymosan is the same in all microwells. Other mixers are
particle phagocytosis incubated in HYPO-IN more laborious to use and temperature is not
since the leukocytes in these samples do not reproducible from one well to the next. The two
phagocytose at all. following shakers work well and are recom-
Assessment of mended:
Phagocyte
Functions by
Flow Cytometry

9.19.18
Supplement 21 Current Protocols in Cytometry
(1) IKA-Minishaker MS 1 (IKA-Works) is MΦs also after phagocytosis. An exception is
not temperature controlled, and was placed in cells from patients with paroxysmal nocturnal
a incubator at 37°C. hemoglobinuria (PNH), who lack CD14. For
(2) iEMS incubator/shaker (Thermo Lab- these patients, phagocytosing MΦs cannot be
systems) contains different shaking levels and discriminated from PMNs by staining for
microtiter plates are temperature controlled. CD14.

Incubation time Digestion


The rate of phagocytosis is astonishingly The measurement of digestion relies on
high. Non-lymphocytes (PMNs + MΦs) fluorescence of liberated targets. Therefore,
phagocytose ∼80 S. aureus bacteria each within target concentrations and incubation times
15 min, and engulf ∼6 to 9 OMV-beads in 7.5 must be chosen so that most of the targets are
min. The phagocytic index increases with in- internalized.
creasing incubation time, but if the incubation Two to three targets adhere to cell nuclei
time is prolonged beyond the point when all after exposure to the detergent NP40 in the
targets are phagocytosed, a diminished rate of Vindeløv’s solution. Because the number of
phagocytosis may be missed. adherent particles is unknown, the fluorescence
When microspheres are used, the phago- of these targets cannot be used to measure
cytic index can be accurately calculated from digestion.
the fluorescence values immediately from the
start of incubation. A short incubation time, Troubleshooting
e.g., 5 min, might seem useful because at that
point the rate of phagocytosis is maximal. Hemolysis
Short incubation times pose problems for Lysis of red blood cells is mandatory since
Maaløe’s method. With short incubation times, red blood cells interfere with measurements by
only a small fraction of the bacteria and zy- blocking the leukocyte access to targets. If the
mosan particles are phagocytosed, leaving only pelleted leukocyte suspension is red after the
a small difference between the initial and final first NH4Cl lysis, lysis should be repeated.
counts of free targets. Such a small difference
between target/phagocyte ratios is hard to Cell clumping
measure by Maaløe’s method, and is quantita- Earlier methods relied on Hank’s buffered
tively unreliable. In the author’s hands, the salt solution supplemented with albumin, or
minimum incubation time with initial tar- other buffers. Buffers containing Ca2+ and
get/phagocyte ratio of 20:1 is 7.5 min. When Mg2+ promote leukocyte aggregation. Cell
target/phagocyte ratio is 10:1, the differences clumping is overcome by washing leukocytes
in the initial ratios and those following phago- in DPBS-GA. Note that Ca2+ and Mg2+ are
cytosis become prominent after 7.5 min. omitted from the solution. Cells can be kept in
DPBS-GA on ice for a few hours without af-
Signal collection fecting phagocyte functions.
A defined collection time minimizes the Cell clumping may occur during phagocy-
drift in fluorochrome distribution and fluores- tosis. If so, the number of aggregates can be
cence intensity between samples. Collect data measured using FS versus SS. The percentage
for 30 sec. Signals are typically recorded from of cell clumps should be the same before and
1500 neutrophils, 100 to 200 monocytes, and after phagocytosis. Results should be discarded
24,000 microspheres. if the fraction of doublets and clumps before
and after phagocytosis is not of the same order
Sample sequence of magnitude.
In order to utterly reduce possible drift,
analyze the contents of two parallel microwell Adhesion to the sample flow lines in the
series in opposite sequence. flow cytometer
Adhesion of targets and leukocytes to the
Discrimination of PMNs from MΦs lines of the sample flow system is minimized
The phagocytosis of microspheres increases by diluting the suspensions in EDTA and by
the SS of MΦs. Therefore, phagocytosing MΦs keeping the samples on ice until the measure-
cannot be discriminated from phagocytosing ment. Microspheres, E. coli, S. typhi, Str.
PMNs by light scatter alone. Staining with epidermidis, S. aureus, and zymosan particles Studies of Cell
antiCD14-PE-Cy5 discriminates PMNs from do not adhere. However, some bacteria, e.g., Function

9.19.19
Current Protocols in Cytometry Supplement 21
meningococci, adhere to sample flow lines. removed by a crude filter followed by sterile
This causes a loss of targets as they pass through filtration (0.22-µm pore size).
the fluidics system, and decreases the measured
target/phagocyte ratio. The phenomenon is ob- Phagocytic index
served as a target/phagocyte ratio higher after This problem regards the differential count
phagocytosis than before. Percentage phagocy- of leukocytes and targets. When the tar-
tosis can still be measured accurately, but not get/phagocyte ratio is >20:1, the relative counts
the phagocytic index. The problem can be less- of leukocytes and targets become inaccurate.
ened and possibly eliminated by diluting the For example, with target%/leukocyte%, one
samples to avoid coincidence, and increasing may have a 96%:4% ratio of 24, a 95%:5% ratio
the sample flow rate. Controls using micro- of 19, and a 94%:6% ratio of 15.7. Thus, a small
scopic counts are required. measured difference in the percentage of targets
from 96% to 94% shifts the (apparent) target
Spontaneous phagocytosis load from 24:1 to 15.7:1. This change in target
Some albumin batches are contaminated, in load profoundly affects phagocyte fluores-
particular, with endotoxin that can elicit phago- cence. Accuracy is increased by lowering the
cytosis and oxidative burst, and affect intracel- ratio of targets to phagocytes. Therefore, the
lular processing. To avoid this problem, pur- determination of the concentration of targets
chase a small amount of albumin, record the should be based not on a single measurement,
batch number, and test the albumin in the ab- but repeated measurements of target/leukocyte
sence of serum to assess unwarranted phagocy- ratios using several target dilutions.
tosis and oxidative burst. Compare phagocyte Using a low target/phagocyte ratio dimin-
FITC fluorescence after the phagocytosis of ishes the strain on the phagocytes, and increases
FITC-labeled targets in the absence and pres- the danger of statistical type 1 error. With small
ence of opsonins. If the batch works as ex- targets such as bacteria and microspheres of
pected, purchase and use this albumin batch. similar size, a target/phagocyte ratio of 20:1 is
useful for many purposes.
Spontaneous oxidative burst
DHR in solution is slowly converted to No phagocytosis
R123. If control neutrophils that have not been Proteins and other chemicals can adhere to
phagocytosing microspheres stain green, indi- various tubes used for reagent storage. Use
cating R123 fluorescence, this staining is prob- polypropylene tubes that do not absorb essen-
ably due to the conversion of DHR to R123 tial ingredients such as opsonins.
before phagocytosis. Make a fresh, new DHR
solution. Anticipated Results
Trypan blue quenching Percentage phagocytosis
Several reports in the literature concern in- The measurement of the percentage of
ability to quench FITC fluorescence with try- phagocytosing leukocytes was introduced by
pan blue. Different batches of trypan blue— Hamburger in 1927, and has since been a pa-
even from the same company source—have rameter of choice (Bassøe et al., 1983). Quad-
been reported to vary in quenching ability. This rant statistics must be set with care to monitor
problem is related not to trypan blue, but to the percentage phagocytosis. Usually, phagocytes
relative concentration of trypan blue to the are at least five times more fluorescent than free
intensity of target FITC staining. Therefore, the targets. Non-phagocytosing leukocytes are less
staining procedure is standardized using a de- fluorescent than individual free targets. There-
fined optical density of the bacteria to be FITC fore, when the vertical quad-stat line is set just
stained. The ability of trypan blue to quench above the fluorescence of the targets, phago-
target FITC fluorescence is easily monitored cytes are clearly discriminated from non-
by flow cytometry. phagocytes. Since non-phagocytosing leuko-
If effective quenching of the targets cannot cytes do not fluoresce at all, the discriminator
be performed, targets should be stained less monitors an all-or-none phagocytosis response.
intensively. The FITC staining protocol given The all-or-none discriminator is useful for
above allows effective quenching of FITC-la- the monitoring of receptor deficiencies and lack
Assessment of beled bacteria and zymosan particles by trypan of receptor function. About 96±3% of periph-
Phagocyte
Functions by blue. Another problem with trypan blue is eral blood neutrophils phagocytose S. aureus,
Flow Cytometry clumps. Trypan blue clumps should be first zymosan particles, and OMV-beads and in

9.19.20
Supplement 21 Current Protocols in Cytometry
HYP, PHS, and REC, respectively. Percentage In the absence of DHR, monocytes do not
phagocytosis drops to ∼70% in REC-IN with fluoresce in the green. When incubated with
OMV-beads as target and is 0% to 5% in HYP- DHR 7.5 min at 37°C, monocytes become
IN and with zymosan particles as target. Per- green fluorescent even in the absence of serum,
centage phagocytosis of S. aureus depends on OMV-beads, mixing, and antiCD14. The per-
the PHS. It varies between 20% and 70% in centage of monocytes in region 1 is 89±9%.
PHS-IN, but is stable for a long time for any AntiCD14 does not influence the monocyte
one PHS batch. This implies that complement R123 fluorescence.
and IgG alone sustain phagocytosis by ∼96±3% The oxidative ratio is a derived parameter
and 70% of the peripheral blood neutrophils. that informs on the conversion of DHR to R123
Percentage phagocytosis by MΦs is ∼70% per ingested bead. MΦs start with a higher
to 75% with S. aureus, zymosan particles, and R123 fluorescence than do neutrophils. With
OMV-beads in HYP, PHS, and REC, respec- increasing incubation time, the monocyte oxi-
tively. The value drops to the order of 5%, 10% dative ratio declines, whereas that of PMNs
to 50%, and 20% in HYP-IN, PHS-IN, and increases. After 15 min, the values are of the
APS, respectively. same order of magnitude in both cell types.
These results are obtained in spite of the
regular observation that all PMNs from the Chemotaxis
same suspensions carry FcγRIII (CD16) and The controls contain lymphocytes, mono-
FcγRII (CD32), and all the MΦs carry CD32 cytes, and neutrophils in proportion to the pe-
and FcγRI (CD64). Under the same conditions ripheral blood counts. Following spontaneous
none of the PMNs carries CD64, and MΦs do migration and chemotaxis, only neutrophils are
not express CD16. Thus, there is an expected observed in the FS versus SS cytograms. Ex-
discrepancy between receptor expression and pected spontaneous migration is ∼30% to 50%,
the ability to phagocytose by the same recep- and chemotaxis 60% to 90%. Chemotaxis val-
tors. ues depend on the concentration and activity of
ZAS and other chemotactic agents (IL-8, fMLP,
Phagocyte subpopulations LTB4, and others).
One advantage of using fluorescent micro-
spheres is their low CV of ∼1.0, as compared Time Considerations
to the CV of FITC-labeled bacteria or zymosan The whole procedure, i.e., Basic Protocols
particles, which is ∼30% to 40%. The latter high 1 to 4, is a demanding 1-day workload. Four
CV makes impossible the accurate discrimina- parallel samples can be investigated in 1 day,
tion of subpopulations of phagocytes differing but it is advisable to select those parts of the
in fluorescence, whereas such a discrimination individual protocols that are suited for the pur-
between cell subpopulations is possible with pose at hand. In Basic Protocol 1, e.g., a suitable
microspheres. bacteria strain can be selected for study, perhaps
When leukocytes and OMV-beads are incu- from a patient, and zymosan particles and mi-
bated with inactive serum, the neutrophils do crospheres can be ignored. Likewise, comple-
not associate with OMV-beads. In REC, nearly ment receptors and serum complement opsonic
all neutrophils are phagocytosing. The percent- activity can easily and rapidly be investigated
age oxidative phagocytosis and the phagocytic by using zymosan particles alone. Micro-
index vary within narrow limits. The difference spheres are useful for the investigation of spe-
between percentage phagocytosis and percent- cific antigens, opsonins, and respiratory burst.
age oxidative burst, i.e., percentage in region 4 However, oxidative burst can also be studied
minus percentage in region 1, is of the order using unstained, non-fluorescent microorgan-
0.6%. The fraction of non-phagocytosing neu- isms and zymosan particles.
trophils is ∼1.5%. The fluorescence of the After the addition of the sample to Vin-
OMV-beads is stable for several years. deløv’s solution, the EB and FITC staining of
In APS, monocytes are mainly localized to targets are stable at least 2 to 3 days in the dark
region 1. In REC, the majority of monocytes at 4°C. Flow cytometric investigations can
are observed in region 2. Very few MΦs are therefore be delayed.
resting, i.e., in region 3.
With APS and in the presence of DHR,
PMNs do not display green fluorescence.
Phagocytosis is triggered in REC, and a single Studies of Cell
population is observed. Function

9.19.21
Current Protocols in Cytometry Supplement 21
Literature Cited Cantinieaux, B., Hariga, C., Courtoy, P., Hupin, J.,
Bassøe, C.-F., Laerum, O.D., Glette, J., Hopen, G., and Fondu, P. 1989. Staphylococcus aureus
Haneberg, B., and Solberg, C.O. 1983. Simulta- phagocytosis. A new cytofluorometric method
neous measurement of phagocytosis and using FITC and paraformaldehyde. J. Immunol.
phagosomal pH by flow cytometry: Role of pol- Methods 121:203-208.
ymorphonuclear neutrophilic leukocyte gran- Hurst, J.K., Albrich, J.M., Green, T.R., Rosen, H.,
ules in phagosome acidification. Cytometry and Klebanoff, S. 1984. Myeloperoxidase-de-
4:254-262. pendent fluorescein chlorination by stimulated
Bassøe, C.-F. 1984. Processing of Staphylococcus neutrophils. J. Biol. Chem. 259:4812-4821.
aureus and zymosan particles by human leuko- Lehmann, A.K., Halstensen, A., Holst, J., and
cytes measured by flow cytometry. Cytometry Bassøe, C.-F. 1997. Functional assays for evalu-
5:86-91. ation of serogroup B meningococcal structures
Bassøe, C.-F. 2000. Flow cytometric quantitation of as mediators of human opsonophagocytosis. J.
phagocytosis in acute myelogenous leukemia. Immunol. Methods 200:55-68.
Acta Haematol. 102:163-171.
Bassøe, C.-F., Smith, I., Sørnes, S., Halstensen, A.,
and Lehmann, A.K. 2000. Concurrent measure- Contributed by Carl-Fredrik Bassøe
ment of antigen- and antibody-dependent oxida- Haukeland University Hospital
tive burst and phagocytosis in monocytes and Bergen, Norway
neutrophils. Meth. Enzymol. 21:203-220.

Assessment of
Phagocyte
Functions by
Flow Cytometry

9.19.22
Supplement 21 Current Protocols in Cytometry

You might also like