You are on page 1of 14

Flow cytometry B3

John F. Dunne and Holden T. Maecker

now bounded more by the complexity of thought


Introduction than by the availability of sophisticated experimen-
tal devices.
The technologies associated with watching cells
flow through an image plane have developed in sev-
eral important directions over the last 30 years. This Mechanistic principles
chapter will focus on flow cytometers,tools that have
optimized fluidics,electronics and optics to generate Fundamentally, flow cytometers measure fluorescent
extraordinary measurement precision and dimen- or scattered light emitted by a cell during its illumi-
sionality on samples of cells in suspension. Outside nation in bright light, typically a highly focused laser
the scope of this discussion remains the continuum beam [2].In most cases,the cell stream is positioned
of devices ranging from real optical microscopes using hydrodynamic focusing,in which a thin core of
that create pictures of cells moving in situ like blood cell suspension is limited to the centre of a larger
cells flowing in a capillary, to simple counting flowing sheath fluid.The cells thus arrive sequential-
devices that monitor the passage of particles in a ly into the laser beam at thousands of cells per sec-
sensing region. ond.
While the fundamental fluidics constraints of Light is scattered by the cells, and this scattered
flow cytometers have not changed, the equipment light is usually measured at narrow angles just above
available to analyze the particles as they pass and below the laser beam (commonly called “FOR-
through a flow cell has been enhanced dramatical- WARD SCATTER”), and by a separate detector at wider
ly with developments in lasers, digital electronics, angles orthogonal to the beam (commonly called
fluorescence chemistries and computer power. “SIDE SCATTER”). FORWARD SCATTER is descriptive of the
Analysis rates have migrated from hundreds to tens size of the cell, while SIDE SCATTER is proportional to
of thousands of cells per second, but more impor- size and granularity. Thus in commonly analyzed
tantly, the number of colours commonly simultane- populations of blood cells, platelets, lymphocytes,
ously measured has gone from one or two in the monocytes and granulocytes can be distinguished
1970s, to four becoming common in the 1990s, to reasonably well simply on the basis of their intrinsic
the eight to twelve colour experiments being pub- light-scattering properties (Fig. 1).
lished recently [1]. These polychromatic experi- More valuably, cells can be stained with fluores-
ments create a new kind of information about flow cent reporter molecules, and the binding of these
samples, as well as a new set of technical chal- reporters can describe extremely subtle phenotypes.
lenges. The optimization of instrument and experi- The most common class of reporters is fluorescently
ment protocols and the efficient analysis of this conjugated monoclonal antibodies (mAb). Thou-
kind of data requires mind-stretching exercises in sands of these products are commercially available.
multidimensional thinking. Useful highly dimen- The antibody protein binds very specifically to par-
sional commercial tools are beginning to enter ticular EPITOPES in proteins present on the cell sur-
research laboratories, and scientists are using them face, or if the cell is permeabilized the antibody can
with some satisfaction.As a result, the technology is enter the cell and bind to EPITOPES within. Since the
184 Flow cytometry

FIGURE 1. DISCRIMINATION OF LEUKOCYTE SUBSETS BY LIGHT SCATTER


The labeled subsets of leukocytes were fluorescently stained with specific mAb (not shown), and the specific mAb-
stained populations were coloured as indicated. These populations were then displayed in a dot plot of forward ver-
sus side scatter. A threshold was set on forward scatter, excluding much of the platelet population, in order to prevent
the collection of small debris that would otherwise interfere with the analysis. Note that the coloured populations are
reasonably well resolved from each other. However, the relative positions of these populations will depend upon the
treatment of the sample (in this example, formaldehyde-fixed and detergent-permeabilized cells were used).

antibody is covalently linked to a fluorescent mole- antigenic protein (see Box 1). By using several mAb,
cule, the antibody binding is directly correlated to each conjugated to fluorescent dyes with character-
the fluorescent signal emanating from the cell dur- istic colours, combinations of antibody binding can
ing laser excitation. In a well-developed assay with characterize populations of cells that may coexist in
binding properties and chemistries well understood, suspension.These and other commonly used fluores-
the brightness associated with antibody binding can cence strategies will be discussed in more detail
be a direct measure of the abundance of the relevant below.
Classification/types of assay 185

BOX 1. WHY USE FLOW CYTOMETRY AS AN ANALYTIC METHOD?

It is possible to use many different methods for quantifying the proteins expressed by a population of cells. In addition
to flow cytometry, Western blotting, immunoprecipitation, ELISA, RIA, enzyme-linked immunospot (ELISPOT), fluores-
cence microscopy, and immunohistochemical staining are all methods used to quantify cellular proteins. Of these, only
flow cytometry, ELISPOT, fluorescence microscopy, and immunohistochemical staining provide information on a single-
cell basis.The others are bulk assays that do not quantify the number or phenotype of cells that are expressing the pro-
tein of interest (though they may provide other information,like the size of the targeted protein).Of the single-cell assays,
flow cytometry is unique in the number of fluorescence parameters that have been combined in a single assay.Thus, it
provides the richest information, on a per-cell basis, of the current generation of assays for cellular protein analysis.

Classification/types of assay rescently conjugated CD4 mAb are added to whole


blood, generally as part of a cocktail of 2 to 4 anti-
Immunophenotype bodies which together uniquely identify those lym-
phocytes that bear the CD4 antigen.With specialized
In the 1980s, with the early propagation of FLOW software tools and sample preparation procedures
CYTOMETRY and mAb,there was some expectation that that are fully validated and approved as in vitro diag-
the protein surfaces of hematopoietic cells could be nostics, the flow cytometer can report the number of
mapped with an ever-increasing complexity to reveal CD4 cells remaining per microlitre of blood. While
changes in the types of cells present or their relative normal ranges can vary around 1000 cells,AIDS pro-
abundance that might correlate with clinical disease gression commonly results in levels that are half of
onset or progression. In the intervening 20 years, with that, and profound disease is usually correlated with
thousands of fluorescent antibodies commercially CD4 counts at or below 200 CD4 cells per µl.
available, the management of only two patient class- While the assay is commonly available to AIDS
es is substantially driven by these kinds of flow practitioners in the developed world, technical and
assays, HIV disease and leukemia/lymphoma. financial barriers have thus far prevented the propa-
gation of the current CD4 assays in environments
with less well-funded medical infrastructures.
Flow immunophenotyping in HIV disease Technology providers are now working with govern-
ments and other funding organizations to develop
It’s now clear that one of the most proximate and dra- viable alternatives and/or equivalents to address this
matic effects of HIV infection is an eventually lethal critical world health issue.
loss of CD4+ T cells [3].This was one of the first easi-
ly recognized manifestations of infection, indeed the
first clinical test that helped identify AIDS patients Flow immunophenotyping in leukemia/
[4], and has been one of the most useful analytical lymphoma
tools available to characterize mechanisms of HIV
pathologies as well as therapeutic benefits of anti- The general observation that the immunopheno-
HIV pharmaceuticals and other treatment modali- types commonly found in peripheral blood samples
ties. More recently augmented by assays that quanti- are relatively stable may have been disappointing in
fy viral load, CD4 counting remains the standard some regards, but it was helpful in that it allows the
method of monitoring disease progression [4]. recognition of unusual phenotypes characteristic of
The CD4 counting assay is relatively simple transformed cells. Especially in the case of trans-
among FLOW CYTOMETRY assays. Most commonly, fluo- formed cells of hematopoietic lineages, these unusu-
186 Flow cytometry

al phenotypes have now been mapped into more or CYTOMETRY to measure the response. One of the most
less broadly recognized categories of malignant dis- common assays of this type is the measurement of
ease,and flow cytometric assays are commonly used CYTOKINE production in lymphocytes in response to
to help diagnose and monitor leukemias and lym- antigenic stimulation. The production of a CYTOKINE
phomas. can be easily measured using well-established
Several clinical research consortia have pub- reagents and protocols, and is a hallmark of lympho-
lished consensus documents describing the gener- cyte function. With multicolour FLOW CYTOMETRY, the
al utility, practical aspects and interpretation of frequency of responsive cells, the amount and kinet-
combinations of markers and their distribution on ics of CYTOKINE production, and the combination of
cancer cells [5–7]. Still, no commercially available various CYTOKINES can very richly describe an
or otherwise standard kit has been approved for immunological response qualitatively and quantita-
diagnostic use, and common practice is for clinical tively.These assays will be discussed in detail below,
centres to use their own discretion to implement and by way of comparison, other functional assays
and validate such assays. Nevertheless, the standard will be addressed briefly (see Box 2).
of care of patients suffering from these malignan-
cies has been improved, sometimes dramatically,
largely based on practitioners’ ability to detect and Proliferation
characterize leukemias and lymphomas by FLOW
CYTOMETRY. Another hallmark of lymphocyte function is cellular
proliferation. Several flow assays have been well
established in the literature that have been correlat-
Immunotoxicology immunophenotyping ed more or less well to traditional 3H-thymidine
incorporation associated with DNA synthesis. Per-
While most common disease states do not change haps the most directly correlated of these is the use
the frequency of normal blood cell components, the of bromodeoxyuridine (BrdU), an orthologue of
toxicity associated with novel pharmacophores or thymidine used by DNA synthase and incorporated
industrial pollutants can have profound effects. in the nuclei of proliferating cells, and detected by
Recent work establishing consensus protocols for FLUOROCHROME-conjugated specific mAb [9]. Anoth-
the evaluation of immunotoxity of such chemical er proliferation assay utilizes the dilution of a cytoso-
entities documents a growing utility for this class of lic dye, commonly carboxyfluorescein diacetate,
assays [8]. The foci of these assays are typically which results from CYTOKINESis [10]. One of the earli-
rodent,dog or non-human primate models,and mAb est flow assays for proliferation, which is still in com-
reagents are less commonly available or less well mon use, is the direct measurement of DNA content
characterized. The normal ranges of various lym- using a quantitative DNA stain such as propidium
phoid cell types, other environmental factors that iodide or the vital dye Hoescht 33258 [11, 12].This is
can influence these ranges, and the magnitude of especially useful for peripheral blood lymphocytes,
changes in these cell types that should be used as which are natively arrested with a 2c DNA content in
sentinels for concern or to prohibit the use of such the G0 phase of the cell cycle, upon stimulation can
chemicals in various exposure scenarios, all repre- enter S phase, and progress to the 4c DNA content
sent ongoing work. typical of G2/M.This doubling of DNA is easily recog-
nized flow-cytometrically, and the percentage of
cells with more than 2c DNA can be roughly related
Functional assays to traditional metrics like the mitotic index.Another
nucleic acid stain, acridine orange, has the unusual
An important additional class of flow assays consists property of shifting its emission spectrum depend-
of those assays that experimentally induce a func- ing on whether it is bound to single- or double-
tional response in cells of interest, then use FLOW stranded nucleic acid (roughly speaking, RNA or
Components/construction of assays 187

BOX 2. DIFFERENT ASSAYS FOR MEASURING T CELL FUNCTION

In this chapter, measurement of intracellular cytokines by flow cytometry is described in some detail as a method for
determining T cell functional responses after short-term stimulation. However, alternative methods are also in common
use. Some are flow cytometry assays, such as MHC-multimer staining [35], or cytokine capture assays [36]; others use
different analytical platforms, such as ELISPOT [37].The main advantage of flow cytometry as an analytical platform is
its multiparameter capability (see Box 1).Of the flow cytometry methods available for measuring T cells in a short-term
assay, MHC-multimer staining is unique in that it measures T cell specificity rather than function, and thus requires no
activation at all. Multimeric forms of MHC molecules with bound antigenic peptides are produced and labeled with a
fluorochrome such as PE.These can then be used to stain T cells (via their T cell receptor) in much the same way as a
fluorochrome-conjugated antibody. However, their use requires a knowledge of the particular peptide and MHC restric-
tion pattern of a T cell immune response; as such, they are not useful for quantifying the overall T cell response to a
pathogen,especially in a MHC-diverse population.Cytokine capture assays,like intracellular cytokine staining,measure
responses without regard to MHC restriction.The cytokine capture assay is especially useful if one wishes to maintain
the cells in a viable state for sorting and further analysis (since it does not require fixation and permeabilization). Still
newer assays, such as measures of granule exocytosis [38], can also be used on viable cells, and add to the armamen-
tarium of tools available for dissecting T cell responses.

DNA) and has been used to simultaneously report ulatory proteins including CYTOKINES, chemokines,
DNA doubling and the increase in mRNA character- growth factors and hormones. Commercially avail-
istic of lymphoid stimulation [13]. able kits range from completely integrated systems
to basic platforms that can be developed for custom
ANALYTE sets, and are used broadly in basic research
Bead matrix immunoassays and drug discovery proteomics projects. No patient
management tools in this class have yet been
Recently bead-based immunoassays have been approved for in vitro diagnostic use as of this writing.
developed for flow cytometric assessment of various
soluble ANALYTES. Each member of a set of beads is
commonly identified by specific fluorescence level Components/construction of assays
and/or size, and acts as an ANALYTE capture platform
using covalently bound specific mAb.The amount of Traditionally, FLOW CYTOMETRY sample preparation
captured ANALYTE is measured using a second anti- and processing has been constrained by the fact that
body specific for an alternate site of the same ANA- flow cytometers were designed to accept tubes, and
LYTE, this second antibody being conjugated to a flu- those tubes were loaded onto the instrument manu-
orescent reporter. Since the various beads can be ally, one at a time. A second constraint came from
recognized independently, and since each captured instrument set-up and data analysis, which were typ-
ANALYTE can be quantified independently, this assay ically time-consuming and required a certain knowl-
format is well suited to multiplexed analysis. Thus edge base. Both of these constraints are now chang-
small volumes of biological fluids (as little as 10 µl of ing with new instrumentation and software. Current
tears, for example [14]) can be inspected for quan- cytometers can often handle racks of tubes that are
tification of soluble proteins or other LIGANDS for automatically run in a walk-away mode. Plate load-
which specific antibody pairs are available.Common ers are also available that can feed samples from 96-
implementations of this strategy include the simulta- well plates directly to the cytometer, again in a walk-
neous quantification of 5–20 different immunomod- away mode.These developments have been comple-
188 Flow cytometry

mented by software that can either: (1) perform data sus surface staining, for example) will also steer the
analysis simultaneously with acquisition; or (2) per- choice of certain FLUOROCHROME conjugates over oth-
form a batch analysis routine that analyzes all of the ers. For a more complete discussion of the issues of
data from an experiment at once. The usefulness of designing multiparameter experiments, see refer-
such analysis routines is further augmented by flexi- ences [18, 19].
ble analysis templates that can accommodate
changing data without repetitive adjustment of set-
tings by the operator [15]. Cell types
The impact of these changes in FLOW CYTOMETRY
hardware and software have opened up the use of FLOW CYTOMETRY can be performed on any cell type
FLOW CYTOMETRY to larger pre-clinical and clinical that can be rendered into a single-cell suspension.
studies that might involve hundreds of specimens. Since blood cells already exist in this state, they have
However, the steps involved in sample preparation been most widely studied by the technique. Howev-
can still be complex, as detailed below. Automation er, adherent cells or tissues can also be analyzed if
of these steps will further allow the use of FLOW they are dissociated from each other and/or their
CYTOMETRY in high-volume settings where it was pre- SUBSTRATE using either enzymatic (e.g., trypsin) or
viously considered too cumbersome. non-enzymatic (e.g., EDTA) treatments. Where possi-
ble, non-enzymatic dissociation protocols are prefer-
able, because they do not cleave cell-surface pro-
Antibodies for cell staining teins that might be TARGETS of the flow cytometric
analysis.
Since the late 1970s, polyclonal antisera have been
increasingly replaced by mAb for most immunologi-
cal applications,including FLOW CYTOMETRY.MAb can Erythrocyte lysis
be produced in unlimited quantities, have better lot-
to-lot reproducibility, and tend to have lower back- Whole blood consists of relatively HOMOGENEOUS ery-
grounds than polyclonal antisera [16]. throcytes, and a much more complex collection of
Another trend in FLOW CYTOMETRY has been the leukocytes. The latter can be stained with FLUO-
increased use of direct FLUOROCHROME conjugates, ROCHROME-conjugated antibodies in the context of
rather than indirect fluorescence analysis using sec- whole blood, but analysis of whole blood is difficult
ond-step antibodies or other reagents that carry the because of the light-scattering properties of the ery-
FLUOROCHROME tag.While indirect staining can some- throcytes, which are so numerous as to obscure the
times amplify the fluorescence signal compared to illumination of the leukocytes. Fortunately, erythro-
direct staining,it may be at the expense of increasing cytes are differentially sensitive to hypotonic lysis,
background fluorescence [17]. Also, multiparameter and can be removed by incubation of blood with
FLOW CYTOMETRY is much more difficult using indirect ammonium chloride in water. Leukocytes are more
staining methods,due to the potential for interaction resistant to osmotic damage than erythrocytes,
between multiple second-step conjugates and the which cannot exclude this salt, and subsequently
primary antibody TARGETS. take up water and are lysed.Typically, whole blood is
Some of the more common FLUOROCHROMES used treated for 10 minutes with a large volume of ammo-
in FLOW CYTOMETRY today are listed in Table 1. Not all nium chloride solution either before or immediately
of these are compatible with all cytometers, or even after staining with FLUOROCHROME-conjugated anti-
with all other FLUOROCHROMEs,as can be seen by their bodies. An alternative to ammonium chloride lysis
excitation and emission spectra (see for example involves hypotonic lysis in the presence of a fixative
www.bdbiosciences.com/spectra). Other considera- (e. g., formaldehyde), whereby the erythrocytes are
tions of relative brightness and compatibility with preferentially lysed while LEUKOCYTES are fixed in a
certain experimental parameters (intracellular ver- single incubation of 10 minutes or so. Solutions for
Components/construction of assays 189

TABLE 1. COMMONLY USED FLUOROCHROMES FOR ANTIBODY-COUPLED FLOW CYTOMETRY

Fluorochrome Type of molecule Typical excitation laser Approximate emission peak

Fluorescein isotyocyanate (FITC) Small organic 488 nm 518 nm


AlexaFluor 488 Small organic 488 nm 518 nm
Phycoerythrin (PE) Protein 488 or 532 nm 574 nm
PE-Texas Red Protein tandem 488 or 532 nm 615 nm
PE-Cy5 Protein tandem 488 or 532 nm 665 nm
Peridinin chlorophyll protein (PerCP) Protein 488 or 532 nm 676 nm
PerCP-Cy5.5 Protein tandem 488 or 532 nm 695 nm
PE-Cy7 Protein tandem 488 or 532 nm 776 nm
Allophycocyanin (APC) Protein 633 nm 659 nm
AlexaFluor 647 Small organic 633 nm 667 nm
AlexaFluor 700 Small organic 633 nm 718 nm
APC-Cy7 Protein tandem 633 nm 784 nm
Pacific Blue Small organic 405 nm 454 nm
AmCyan Protein 405 nm 487 nm

this procedure are commercially available. After By underlaying whole blood (usually diluted 1:1 with
lysis/fixation, the cells may be directly analyzed by buffer or tissue culture media) with a similar volume
FLOW CYTOMETRY (“no-wash” assays); or they may be of a FICOLL solution, a density gradient is formed.This
subjected to washing to remove the residual red cell biphasic solution is then subjected to centrifugation
debris and unbound fluorochrome-conjugated anti- (typically at about 400 x G for 15-20 minutes). Erythro-
bodies before analysis (“washed” assays). Washed cytes and granulocytes, which have the greatest buoy-
assays typically allow better signal-to-noise discrimi- ant density, are pelleted to the bottom of the FICOLL
nation of fluorescently stained populations, and bet- layer. Lymphocytes and monocytes, which are less
ter visualization of lymphocyte scatter properties. dense, collect at the interface of the plasma and
However, no-WASH-ASSAYS have become popular in FICOLL,where they are collected by pipetting.Platelets,
the clinical marketplace because of their simplicity; the least dense leukocytes, will remain in the plasma
and by detecting cells based on threshold staining layer. Successive centrifugation of the mononuclear
for a common leukocyte antigen,such as CD45,these cells collected from the interface is then carried out
assays can adequately resolve subpopulations of at 250 × G to remove residual FICOLL and further
lymphocytes. deplete the mononuclear cells of platelets.
Density gradient separation techniques are a stan-
dard, albeit time-consuming method for the isolation
Density gradient separation of mononuclear cells from small to large volumes of
blood. However, simpler alternatives are also available
As an alternative to erythrocyte lysis of whole blood, that consist of a gel matrix pre-dispensed into a blood
mononuclear LEUKOCYTES (lymphocytes and mono- collection tube or centrifuge tube. By adding whole
cytes) can be directly isolated from whole blood prior blood and centrifuging at a prescribed speed, the
to staining. Solutions of high molecular weight carbo- mononuclear cell layer can be collected from the top
hydrates, such as FICOLL, are used for this separation, of the gel interface, while erythrocytes and granulo-
which is accomplished on the basis of density [20]. cytes are forced through the gel plug. Such systems
190 Flow cytometry

are available from commercial vendors, and provide hyde-based buffer) and then permeabilized (usually
generally equivalent results with greater convenience with a detergent-based buffer). Commercial reagents
than the FICOLL separation technique [21]. containing fixatives and detergents are readily avail-
able and allow for reproducible fixation and perme-
abilization protocols.
Activation

As described briefly above, functional assays are Cell staining


those in which the cell types of interest are stimulat-
ed in vitro and allowed to manifest some kind of For activation assays designed to measure CYTOKINE
response, the quantity and quality of which is meas- production,intracellular staining is required; but cell-
ured in the flow cytometer. Lymphocyte activation is surface staining is usually also performed in the
a particularly valuable class of functional assays,and same sample to allow phenotyping of the respond-
typically requires specific antigen or polyclonal ing cell population. Depending upon the antibodies
MITOGEN as a stimulus. A commonly measured and EPITOPES targeted, this surface staining can some-
response to this stimulus is CYTOKINE production, times be done together with the intracellular
which can be detected in a time period of as little as CYTOKINE staining, if the targeted EPITOPES are resistant
4 hours (for IFN-γ,IL-2,TNF-α,and IL-4) [22].Visualiza- to the conditions of fixation and permeabilization
tion of CYTOKINE production is accomplished with the [22, 26]. If they are not, a surface staining step is car-
aid of a secretion inhibitor, such as brefeldin A [23] ried out after activation but before fixation and per-
or monensin [24], which allows for intracellular meabilization.
accumulation of the CYTOKINES where they can be For most EPITOPES, surface and/or intracellular
specifically stained after cell permeabilization. Typi- staining can be done by incubation with a cocktail
cal antigen-activation protocols involve 6 hours of of FLUOROCHROME-conjugated antibodies for 30–60
stimulation in the presence of brefeldin A (for pep- minutes at room temperature.Titration of antibodies
tides or MITOGENS),or 6 hours of stimulation with pro- is required for optimal staining (although many man-
tein antigens, the last 4 hours in the presence of ufacturers offer pre-titred antibodies and cocktails of
brefeldin A [25]. The latter protocol allows for pro- antibodies). The optimal titre for surface staining
teins to be processed and presented by MHC mole- (unfixed cells) is often different from that for intra-
cules prior to inhibition of the secretory pathway by cellular staining (after fixation and permeabiliza-
brefeldin A [26].Activation is carried out at 37 °C,but tion).
can be terminated by cooling the cells prior to fur- As noted above, simple surface staining of whole
ther processing [22]. Thus, programmeable water blood can be done using a no-WASH-ASSAY format.But
baths or similar devices can be used to automatical- more complex assays, such as intracellular CYTOKINE
ly time the activation process, even if it is initiated at staining, require washing, both after fixation and per-
the end of a workday. meabilization steps, and after antibody staining.
Because activation of lymphocytes can induce
adhesion molecule expression, a brief treatment
with EDTA at the end of the activation period is rec- Increasing automation and throughput
ommended to dislodge adherent cells from the cul-
ture vessel [26]. The relative complexity of sample preparation, espe-
cially for the more complex functional assays, has
inhibited their application to studies of large num-
Fixation/permeabilization bers of animals or large clinical trials. However,
robotic sample preparation devices are available
For staining of intracellular EPITOPES such as CYTO- and more are in development that would significant-
KINES, cells are first fixed (usually with a formalde- ly aid in this process. For example, robotic worksta-
Components/construction of assays 191

tions are commercially available that can aliquot cells within those gates.Typically, viable cells of inter-
whole blood into staining tubes, add antibodies, est are first identified by their light scatter properties,
incubate, and add erythrocyte lysis buffer. There are using forward and SIDE SCATTER parameters. This may
also workstations that can perform cell washing, and be followed by gating on subsets of lymphocytes, for
thus can automate most of the steps of a washed example, CD3+ (T cells), CD19+ (B CELLS), etc. Subsets
assay. Integration of these robotic workstations can of T cells (CD4+,CD8+) may be identified through fur-
not only increase the number of samples handled in ther gating. In the case of functional assays, a respon-
these applications, but can also lend considerable sive population, e.g., CYTOKINE-positive, is quantified
standardization to processes that otherwise are high- from the subset of interest, e.g., CD8+ T cells.
ly operator-dependent. The PRECISE placement of fluorescence and light
Truly high-throughput sample processing, howev- scatter gates is historically done by eye, based upon
er, is best accomplished with multiwell plates. Both recognition of typical patterns of staining (negative,
phenotypic and functional assays can be performed dim, bright, etc.). However, cluster-finding algorithms
in 96-well plates; in fact, protocols for activation, pro- are available in some current FLOW CYTOMETRY soft-
cessing, and analysis of samples for intracellular ware programmes that allow these gates to be drawn
CYTOKINE staining in a single 96-well plate have been in a semi-automated way, and to be responsive to
published [15]. With the availability of plate-based changes from one data file to another [15]. Such
loaders for flow cytometers, much larger numbers of changes might include slight differences in staining
samples can be processed in a single run, with mini- intensities in different donors, between different
mal incremental technician time. Further automa- CYTOKINE antibodies used, etc. By using such cluster-
tion of plate-based sample processing can also be finding algorithms, a gating template can be con-
accomplished using robotic workstations. For exam- structed that can then process all the data of a given
ple, a robotic workstation has been described that experiment without requiring visual checking of
integrates a plate-based liquid handling robot (for each data file. The ungated “raw” data is stored, and
dispensing cells and reagents) with an incubation the gating template can be applied to the raw data
station, plate-based indexing centrifuge, and a flow files either contemporaneously with data acquisi-
cytometer equipped with a plate loader [27]. tion, or in a batch analysis mode at the end of data
Transfer of plates to and from the various stations is acquisition. If data analysis strategies change as a
done with a robotic arm, and software controlling study unfolds, the raw data can be subsequently
the robotic arm also integrates the various compo- reprocessed through revised gating templates. This
nents. Further development of such systems will requires considerably less operator time than tradi-
broaden the ability to use FLOW CYTOMETRY for highly tional methods of data analysis in which the user
parallel studies potentially involving thousands of places gates manually and checks/adjusts them for
samples. each data file, then has to re-do all the work manual-
ly if strategies change.
A final component of data analysis that is often
Data acquisition and analysis overlooked is the incorporation of flow cytometric
data into a database that may also link it with other
Although sample preparation can be made parallel types of measurements (patient clinical data, etc.).
using multiwell plates and automation, sample Fundamentally, this requires the ability to extract
acquisition is still a serial process. Fortunately, tube- measurements of interest from the FLOW CYTOMETRY
and plate-loaders are available that can automate data files into a spreadsheet along with any annota-
acquisition, and software can be used to annotate tion associated with the data files.This can be done
data files before they are run. with modern FLOW CYTOMETRY software packages,
Data analysis involves the setting of “gates”or regions such that manual entry of data into a spreadsheet is
in one- or two-dimensional data space, followed by not necessary.This is a fundamental requirement for
further analysis of the fluorescence properties of
192 Flow cytometry

FIGURE 2. CD4 T CELL ENUMERATION ASSAY


Cells and counting beads are successively gated as described in the text, and the percentages and absolute counts of
various lymphocyte subsets are automatically reported by the analysis software.

any large studies using FLOW CYTOMETRY as one of on a four-colour flow cytometer. An ACQUISITION
their components. THRESHOLD was set on CD45 fluorescence, in order to
identify leukocytes. All cells above this threshold
were collected, with acquisition halted at 20,000
Examples and their application CD45+ events.
Analysis of this assay was done as follows.An ini-
Immunophenotyping assays tial gate was set on CD3+ cells, in order to identify all
T cells. CD4+ and CD8+ T cells were then quantified
An example of an IMMUNOPHENOTYPING assay to quan- from a plot gated on CD3+ cells.Note that this sample
tify T cell subsets is shown in Figure 2. Note that this also contained counting beads which are identified
was done as a “no-wash” assay in four colours, along by very high fluorescence in all colours. Because a
with fluorescent beads to allow absolute counting. known number of these beads was added to a
Fifty microlitres of whole blood were stained with known volume of the blood sample, a simple calcu-
antibodes to CD45, CD3, CD4, and CD8. The blood lation can convert the percentages of each cell sub-
was subjected to fixation/erythrocyte lysis, then run population to an absolute count of cells per micro-
Examples and their application 193

litre of blood. Since percentages are relative to other these gates were set using a cluster-finding algorithm
populations, absolute counts have become a stan- that allows the gate to automatically shift with the
dard readout for reporting these types of results. population from sample to sample [15]. An excep-
Commercial software packages can perform these tion is the final region quantifying IFN-γ+CD69+ cells,
calculations in an automated fashion for this partic- which was a user-defined rectangle, but which was
ular application. “tethered” to the negative population, so that it too
would shift in response to changes in the back-
ground level of fluorescence from sample to sample.
Functional assays In this way,even very rare populations of IFN-γ+CD69+
cells could be quantified,whether they formed a rec-
An example of a functional assay (identifying intra- ognizable cluster or not.
cellular CYTOKINE production) is shown in Figure 3. If desired, one could combine the information
This assay was done by stimulation of whole blood from the above two assays, in order to express the
with peptides derived from human cytomegalovirus HCMV pp65-responsive CD3+CD4+ or CD3+CD4– cells
(HCMV),a common herpesvirus that causes chronic, as an absolute number of cells per microlitre.Again,
latent infection of various tissues. HCMV causes non- this might provide more standardized results in pop-
pathological infection in immunocompetent hosts, ulations that have varying numbers of CD4 and CD8
but is an opportunistic pathogen in immunocompro- T cells (such as HIV patients).
mised hosts [28]. What is the usefulness of identifying and quantify-
The peptides used for stimulation in this assay ing intracellular CYTOKINE responses? One major
were a mixture of 138 15-mers, overlapping by 11 application is the development of new vaccines (see
amino acid residues each, and spanning the Chapter C.1) that are designed to elicit cellular
sequence of the pp65 glycoprotein of HCMV. Pp65 is immunity [30, 31]. These include both prophylactic
an immunodominant protein which contains EPI- and therapeutic vaccines for HIV, cancer, and certain
TOPES that stimulate CD4 and CD8 T cell responses in other viral and intracellular bacterial pathogens.
a variety of HLA haplotypes [29]. By using such over- Establishing biomarkers of immunogenicity of vac-
lapping peptides,both CD4 and CD8 T cell responses cines (and eventually surrogate markers of protec-
can be stimulated with relative efficiency [25]. The tion) would greatly facilitate the comparison of differ-
blood was stimulated with this peptide mixture for ent vaccine constructs and strategies,and allow more
six hours in the presence of brefeldin A, followed by rapid improvement of vaccines for these diseases.
EDTA treatment, fixation/erythrocyte lysis, permeabi- In the area of immunotoxicology (see Chapter
lization, and surface/intracellular staining. The anti- D.1), quantification of functional T cell responses,
bodies used included anti-IFN-γ,CD69,CD4,and CD3. whether to specific antigens or to MITOGENS, could
Because this was a washed assay, it was not neces- also provide valuable information [30]. While
sary to use a reagent like CD45 for fluorescence trig- IMMUNOPHENOTYPING can potentially uncover gross
gering. Instead, a threshold was set on FORWARD SCAT- changes in cellular subsets in response to a drug,
TER to eliminate small debris,and an acquisition gate functional assays can uncover much more subtle
was set around the cluster of small lymphocytes in changes. Suppression (or augmentation) of antigen
forward versus SIDE SCATTER. Acquisition was stopped or MITOGEN responses could give important clues to
when 40,000 CD4+ T cells were collected. immunological side-effects of drugs. This could be
Analysis of this assay was done as follows.An ini- done as part of the early screening of drug candi-
tial gate was set on CD3+ lymphocytes in a CD3 ver- dates, before expensive animal studies are undertak-
sus SIDE SCATTER plot. Secondary gates were set on en. Functional FLOW CYTOMETRY assays could also be
CD3+CD4+ and CD3+CD4– cells, respectively. From used to monitor the dosing of immunomodulatory
each of the latter gates, plots of CD69 versus IFN-γ drugs in settings such as transplantation and autoim-
expression were displayed,and cells positive for both munity [32, 33]. By standardizing, automating, and
of these activation markers were quantified. All of increasing the ease and throughput of these assays,
194 Flow cytometry

FIGURE 3. CYTOKINE FLOW CYTOMETRY ASSAY


(A) Cells are initially gated to identify CD3+CD4+ and CD3+CD4– lymphocytes (the latter population being substan-
tially equivalent to CD8+ T cells). (B) Functional responses from each of these populations are then quantified as the
percentage of the gated population that is positive for both CD69 (a surface marker of activation) and IFN-γ (a T cell
cytokine). The response from an antigen-stimulated sample (top plots) is then compared to that from a control, unstim-
ulated sample (bottom plots). Note that even a very low percentage of positive cells, e.g., 0.1%, can be easily differ-
entiated from the control, when the percentage of positive cells in the control sample is low enough, as in these exam-
ples. The statistical significance of the difference between control and test samples can also be calculated [34].
References 195

they can become ever more potent tools for on the immunophenotypic analysis of hematologic
immunological research, immunotoxicology, and neoplasia by flow cytometry: medical indications.
clinical medicine. Cytometry 30: 249–263
6 Stewart CC, Behm FG, Carey JL, Cornbleet J, Duque RE,
Hudnall SD, Hurtubise PE, Loken M,Tubbs RR,Worms-
Summary ley S (1997) U.S.-Canadian Consensus recommenda-
tions on the immunophenotypic analysis of hemato-
FLOW CYTOMETRY is a powerful technique for analyz- logic neoplasia by flow cytometry: selection of anti-
ing cells in suspension, and has been extensively body combinations. Cytometry 30: 231–235
applied to the analysis of lymphocytes and their sub- 7 Rothe G, Schmitz G (1996) Consensus protocol for the
sets. Flow cytometric assays can be divided into flow cytometric immunophenotyping of hematopoiet-
IMMUNOPHENOTYPING assays and functional assays,the ic malignancies. Working Group on Flow Cytometry
latter requiring in vitro stimulation of cells in order to and Image Analysis. Leukemia 10: 877–895
read out a response, such as CYTOKINE production. 8 Burchiel SW, Kerkvliet NL, Gerberick GF, Lawrence DA,
Such assays are being applied to the monitoring of Ladics GS (1997) Assessment of immunotoxicity by
clinical disease states and responses to vaccination multiparameter flow cytometry. Fundam Appl Toxicol
or immunomodulation,and they have great potential 38: 38–54
to be used in measuring immunotoxicology as well. 9 Mehta BA,Maino VC (1997) Simultaneous detection of
DNA synthesis and cytokine production in staphylo-
coccal enterotoxin B activated CD4+ T lymphocytes by
Selected readings flow cytometry. J Immunol Methods 208: 49–59
10 Lyons AB, Parish CR. (1994) Determination of lympho-
Givan AL (2001) Flow cytometry: First Principles. J. Wiley, cyte division by flow cytometry. J Immunol Methods
New York 171: 131–137
Robinson JP (ed) (1999) Current Protocols in Cytometry 11 Braylan RC, Diamond LW, Powell ML, Harty-Golder B
(Current Protocols Series). J.Wiley, New York (1980) Percentage of cells in the S phase of the cell
Purdue cytometry bulletin board: http://www.cyto.purdue. cycle in human lymphoma determined by flow
edu/hmarchiv/cytomail.htm (Accessed November cytometry. Cytometry 1: 171–174
2004) 12 Taylor IW, Milthorpe BK (1980) An evaluation of DNA
fluorochromes, staining techniques, and analysis for
flow cytometry. I. Unperturbed cell populations. J His-
References tochem Cytochem 28: 1224–1232
13 Darzynkiewicz Z (1994) Simultaneous analysis of cel-
1 Herzenberg LA, Parks D, Sahaf B, Perez O, Roederer M lular RNA and DNA content. Methods Cell Biol 41:
(2002) The history and future of the fluorescence acti- 401–420
vated cell sorter and flow cytometry: a view from Stan- 14 Cook EB, Stahl JL, Lowe L, Chen R, Morgan E,Wilson J,
ford. Clin Chem 48: 1819–1827 Varro R,Chan A,Graziano FM,Barney NP (2001) Simul-
2 Givan AL (2001) Flow cytometry: First Principles. J. taneous measurement of six cytokines in a single sam-
Wiley, New York ple of human tears using microparticle-based flow
3 Libman H, Makadon HJ (eds) (2003) HIV (2nd edn). cytometry: allergics vs. non-allergics. J Immunol Meth-
American College of Physicians, Philadelphia ods 254: 109–118
4 Mandy F, Nicholson J,Autran B, Janossy G (2002) T-cell 15 Suni MA, Dunn HS, Orr PL, deLaat R, Ghanekar SA,
subset counting and the fight against AIDS: reflections Bredt BM, Maino VC, Maecker HT (2003) Performance
over a 20-year struggle. Cytometry 50: 39–45 of plate-based cytokine flow cytometry with automat-
5 Davis BH, Foucar K, Szczarkowski W, Ball E, Witzig T, ed data analysis. BMC Immunology 4: 9
Foon KA, Wells D, Kotylo P, Johnson R, Hanson C et al 16 Kelley KW, Lewin HA (1986) Monoclonal antibodies:
(1997) U.S.-Canadian Consensus recommendations
196 Flow cytometry

pragmatic application of immunology and cell biolo- compromised humans. A review. Dermatology 200:
gy. J Anim Sci 63: 288–309 189–195
17 Zolla H (1999) High-sensitivity immunoflourescence/ 29 Kern F, Bunde T, Faulhaber N, Kiecker F, Khatamzas E,
flow cytomtery: Detection of cytokine receptors and Rudawski IM, Pruss A, Gratama JW, Volkmer-Engert R,
other low-abundance membrane molecules. In: JP Ewert R et al (2002) Cytomegalovirus (CMV) phos-
Robinson (ed): Current Protocols in Cytometry. Unit phoprotein 65 makes a large contribution to shaping
6.3. J.Wiley, New York the T cell repertoire in CMV-exposed individuals. J
18 Baumgarth N, Roederer M (2000) A practical Infect Dis 185: 1709–1716
approach to multicolor flow cytometry for 30 Maecker HT, Maino VC, Picker LJ (2000) Immunofluo-
immunophenotyping. J Immunol Methods 243: 77–97 rescence analysis of T-cell responses in health and dis-
19 De Rosa SC, Brenchley JM, Roederer M (2003) Beyond ease. J Clin Immunol 20: 391–399
six colors: a new era in flow cytometry. Nat Med 9: 31 Maecker HT, Maino VC (2003) T cell immunity to HIV:
112–117 defining parameters of protection. Current HIV
20 Coligan JE, Kruisbeek AM, Margulies DH, Shevach EM, Research 1: 249–259
Strober W (eds) (1995) Current Protocols in Immunol- 32 Sindhi R,Allaert J,Gladding D,Koppelman B,Dunne JF
ogy.Wiley, London (2001) Cytokines and cell surface receptors as target
21 Nomura LE,DeHaro ED,Martin LN,Maecker HT (2003) end points of immunosuppression with cyclosporine
Optimal preparation of rhesus macaque blood for A. J Interferon Cytokine Res 21: 507–514
cytokine flow cytometric analysis. Cytometry 53A: 33 Sindhi R, Allaert J, Gladding D, Haaland P, Koppelman
28–38 B, Dunne J, Sehgal S (2003) Modeling individual varia-
22 Nomura LE, Walker JM, Maecker HT (2000) Optimiza- tion in biomarker response to combination immuno-
tion of whole blood antigen-specific cytokine assays suppression with stimulated lymphocyte responses-
for CD4(+) T cells. Cytometry 40: 60–68 potential clinical implications. J Immunol Methods
23 Waldrop SL, Pitcher CJ, Peterson DM, Maino VC, Picker 272: 257–272
LJ (1997) Determination of antigen-specific memo- 34 Motulsky H. (1995) Intuitive Biostatistics. Oxford:
ry/effector CD4+ T cell frequencies by flow cytometry: Oxford Univ. Press, 197-203.
evidence for a novel, antigen-specific homeostatic 35 Altman JD, Moss PAH, Goulder PJR, Barouch DH,
mechanism in HIV-associated immunodeficiency. J McHeyzer-Williams MG, Bell JI, McMichael AJ, Davis
Clin Invest 99: 1739–1750 MM. (1996) Phenotypic analysis of antigen-specific T
24 Prussin C, Metcalfe DD (1995) Detection of intracyto- lymphocytes. Science 274: 94–96
plasmic cytokine using flow cytometry and directly 36 Brosterhus H, Brings S, Leyendeckers H, Manz RA, Mil-
conjugated anti-cytokine antibodies. J Immunol Meth- tenyi S, Radbruch A,Assenmacher M, Schmitz J (1999)
ods 188: 117–128 Enrichment and detection of live antigen-specific
25 Maecker HT, Dunn HS, Suni MA, Khatamzas E, Pitcher CD4(+) and CD8(+) T cells based on cytokine secre-
CJ,Bunde T,Persaud N,Trigona W,Fu TM,Sinclair E et al tion. Eur J Immunol 29: 4053–4059
(2001) Use of overlapping peptide mixtures as anti- 37 Hutchings PR, Cambridge G,Tite JP, Meager T, Cooke A
gens for cytokine flow cytometry. J Immunol Methods (1989) The detection and enumeration of cytokine-
255: 27–40 secreting cells in mice and man and the clinical appli-
26 Suni MA, Picker LJ, Maino VC (1998) Detection of anti- cation of these assays. J Immunol Methods 120: 1–8
gen-specific T cell cytokine expression in whole blood 38 Betts MR, Brenchley JM, Price DA, De Rosa SC, Douek
by flow cytometry. J Immunol Methods 212: 89–98 DC, Roederer M, Koup RA (2003) Sensitive and viable
27 Dunne JF, Maecker HT (2004) Automation of cytokine identification of antigen-specific CD8+ T cells by a
flow cytometry assays. J Assoc Lab Automation 9: 5–9 flow cytometric assay for degranulation. J Immunol
28 Drago F, Aragone MG, Lugani C, Rebora A (2000) Methods 281: 65–78
Cytomegalovirus infection in normal and immuno-

You might also like