Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Background

The evaluation of sputum leukocytes by flow cytometry (FCM) is an opportunity to assess characteristics of cells residing in the central airways, yet it is hampered by certain inherent properties of sputum including mucus and large amounts of contaminating cells and debris.

Objective

To develop a gating strategy based on specific antibody panels in combination with light scatter properties for flow cytometric evaluation of sputum cells.

Methods

Healthy and mild asthmatic volunteers underwent sputum induction. Manually selected mucus "plug" material was treated with dithiothreitol, filtered and total leukocytes acquired. Multicolor FCM was performed using specific gating strategies based on light scatter properties, differential expression of CD45 and cell lineage markers to discriminate leukocytes from squamous epithelial cells and debris.

Results

The combination of forward scatter and CD45 expression reliably segregated sputum leukocytes from contaminating squamous epithelial cells and debris. Overlap of major leukocyte populations (neutrophils, macrophages/monocytes) required the use of specific antibodies (e.g. CD16, CD64, CD14, HLA-DR) that differentiated granulocytes from monocytes and macrophages. These gating strategies allowed identification of small populations of eosinophils, CD11c+ myeloid dendritic cells, B-cells and natural killer cells.

Conclusions

Multicolor FCM can be successfully applied to sputum samples to identify and characterize leukocyte populations residing on the surfaces of the central airways.

Clinical relevance

This research describes detailed methods to overcome difficulties associated with FCM of sputum samples, which previously has been lacking in the literature. FCM of sputum samples can provide valuable information on inflammation and immunological response elements in the bronchial airways for both clinical diagnostic and research applications and can be a useful tool in inhalation toxicology for assessing health effects of inhaled environmental pollutants.

Free full text 


Logo of nihpaAbout Author manuscriptsSubmit a manuscriptHHS Public Access; Author Manuscript; Accepted for publication in peer reviewed journal;
Inhal Toxicol. Author manuscript; available in PMC 2013 Jun 9.
Published in final edited form as:
PMCID: PMC3677049
NIHMSID: NIHMS317901
PMID: 21639708

Flow cytometry of sputum: assessing inflammation and immune response elements in the bronchial airways

John C. Lay, PhD, DVM,1,2 David B. Peden, MD, MS,1,2,3 and Neil E. Alexis, PhD1,2

Abstract

Background

The evaluation of sputum leukocytes by flow cytometry is an opportunity to assess characteristics of cells residing in the central airways, yet it is hampered by certain inherent properties of sputum including mucus and large amounts of contaminating cells and debris.

Objective

To develop a gating strategy based on specific antibody panels in combination with light scatter properties for flow cytometric evaluation of sputum cells.

Methods

Healthy and mild asthmatic volunteers underwent sputum induction. Manually selected mucus “plug” material was treated with dithiothrietol, filtered and total leukocytes acquired. Multicolor flow cytometry was performed using specific gating strategies based on light scatter properties, differential expression of CD45 and cell lineage markers to discriminate leukocytes from squamous epithelial cells and debris.

Results

The combination of forward scatter and CD45 expression reliably segregated sputum leukocytes from contaminating squamous epithelial cells and debris. Overlap of major leukocyte populations (neutrophils, macrophages/monocytes) required the use of specific antibodies (e.g. CD16, CD64, CD14, HLA-DR) that differentiated granulocytes from monocytes and macrophages. These gating strategies allowed identification of small populations of eosinophils, CD11c+ myeloid dendritic cells, B cells and NK cells.

Conclusions

Multicolor flow cytometry can be successfully applied to sputum samples to identify and characterize leukocyte populations residing on the surfaces of the central airways.

Keywords: induced sputum, flow cytometry, immunophenotype, methods, human

INTRODUCTION

Flow cytometry (FCM) is a useful tool for a variety of both clinical diagnostic and research applications and is used extensively for immunophenotyping blood. The rationale for its application to sputum is based on the fact that induced sputum techniques provide a mechanism for procuring viable cells derived from surfaces of central airways (Alexis et al. 2001b), a region of the lung which is difficult to sample non-invasively in humans. While sputum induction retrieves sufficient cells for FCM, its application to sputum samples remains relatively novel, despite its potential for yielding valuable information on the functional and phenotypic characteristics of inflammatory and immune cells residing on bronchial surfaces.

Unlike blood and bronchoalveolar lavage (BAL) samples, sputum contains variable amounts of mucus, contaminating epithelial cells and endogenous or exogenous debris which can render FCM analysis challenging, likely causing reluctance by many investigators to use it for evaluating sputum. As a result, BAL cells are often used as surrogates for cells residing in the central airways; however, this is less than optimal because these techniques sample different regions of the lung (central airways vs. alveolar parenchyma) and the leukocyte composition (Table 1) is quite different between the two. (Balbi et al. 2007; Bienkowska-Haba et al. 2002; Pizzichini et al. 1998; Grootendorst et al. 1997; Alexis et al. 2000) BAL and sputum cells also possess different functional and phenotypic characteristics. (Alexis et al. 2000) For these reasons, application of FCM to sputum samples (as opposed to BAL) is, in our view, preferable and, perhaps, more appropriate for assessing inflammation of the bronchial airways.

Table 1

Comparison of Leukocyte Populations of Blood, Sputum and BALF in Healthy Individuals.*

Cell typeBloodSputumBALF
Monos/Macs2 – 10% (monos)30 – 95%75 – 95%
Dendritic cells0.5 – 1.0%0.5 – 1.5%0.5 – 1.0%
Neutrophils40 – 75%5 – 95 %0 – 1%
Eosinophils1 – 6%0 – 1%0 – 2%
Lymphocytes20 – 40%0 – 3%0 – 23%
Bronchial Epith.NA0 – 10%0 – 3%
Squamous Epith.NA1 – 60%Rare
Basophils0 – 1%Extremely RareExtremely Rare
Mast cells0%0%0%
Macrophages are absent from blood. The values for sputum and BALF include monocytes and macrophages combined.

Early studies, (Alexis et al. 2001a; Alexis et al. 2000) as well as some more recent FCM studies from other labs were somewhat technically limited in that they relied primarily on light scatter properties for gating sputum populations and used only two fluorochromes (FITC and PE) for analysis of sputum. Thus anti-CD45, a critical marker for differentiating leukocytes from debris was generally not used. As a result, the purity of gated leukocyte populations in these studies likely suffered. Some studies have used specific cell lineage markers (e.g.: CD3, CD4, CD14, HLA-DR or other markers) to identify and gate specific cells of interest (Barry et al. 2002; Barry and Janossy2004; Frankenberger et al. 2004; Janossy et al. 2008; Holden et al. 2008; McCarthy et al. 2007) and a few more recent studies (Leckie et al. 2003; Dominguez et al. 2004; Jaksztat et al. 2004; Lay et al. 2007; Alexis et al. 2009; Antoniou et al. 2005; Dua et al. 2010) employed more intricate approaches; however, none have provided detailed descriptions of a comprehensive and rigorous approach using FCM for differentiating sputum leukocytes from debris.

Here we describe strategies using a combination of light scatter properties, CD45 expression, and differential expression of lineage-specific cell surface markers which allow more confident identification of sputum leukocytes. Antibody combinations were designed to assess expression of specific cell surface proteins associated with innate and adaptive immunity. Surface proteins associated with innate immune function include Fc-gamma receptors (FcγRI/CD64 and FcγRIII/CD16), the LPS receptor (CD14) and complement receptors (CR3/CD11b; CR4/CD11c), as well as Toll-like-receptors 2 and 4 (CD282 and CD284, respectively) and the low- and high-affinity IgE receptors (CD23/FcεR-IIα and FcεRIα, respectively). These are found primarily on mononuclear phagocytes and granulocytes and to lesser degree on other leukocytes. While they function primarily in the innate immune system, their functions also overlap with adaptive immune responses. Proteins associated primarily with adaptive immune responses include HLA-DR/MHC-II, co-stimulatory molecules CD80/B7.1, CD86/B7.2, CD40 and CD83 (tubes 4, 5 and 6), which are expressed primarily on antigen presenting cells (APC) including DCs, monocytes, macrophages and lymphocytes. They function in antigen presentation and activation of T-cells and B-cells to elicit antigen specific immune responses and modulate immune-mediated inflammation.

The methods described here provide detailed guidance to investigators in both research and clinical settings who wish to use FCM for evaluating sputum samples.

METHODS

Extended descriptions and discussion of methods presented here are included in the Appendix.

Subjects and IRB approval

Induced sputum was obtained from both healthy and mild allergic asthmatic or allergic non-asthmatic volunteers under our screening protocol which was reviewed and approved by the University of North Carolina Committee on the Rights of Human Subjects (Institutional Review Board). Asthmatic volunteers had mild symptoms of asthma as defined under section 3 of the 2007 NHLBI guidelines for the diagnosis and management of asthma and all had a positive methacholine challenge test (PC20 ≤ 10 mg/ml), defined as a 20% decrease from the baseline forced expiratory volume in the first second (FEV1) following an inhaled provocative challenge dose of ≤ 10mg methacholine/ml. Atopy was demonstrated by a positive immediate skin test response to one of the following allergen mixes: 2 species of house dust mite (Dermatophagoides farinaea and Dermatophagoides pteronyssinus), cockroach, tree mix, grassmix, weedmix, moldmix1, moldmix2, rat, mouse, guinea pig, rabbit, cat or dog.

Sample Collection and Processing

Sputum induction and processing techniques are based on the those of Hargreave et al (Hargreave et al. 1998) and are detailed elsewhere. (Alexis et al. 2000) Briefly, only the cell-enriched mucus “plugs” manually selected from the surrounding clear fluid were processed to minimize squamous cell contamination and dilution from saliva. (Pizzichini et al. 1996; Spanevello et al. 1998; Alexis et al. 2006) Sputum samples should immediately be placed on ice and processed within 2 hours.(Efthimiadis et al. 2002a) Excessive or prolonged exposure to dithiothrietol (DTT) during processing adversely affects both cell viability and surface marker expression (Efthimiadis et al. 1997; Loppow et al. 2000) and should be avoided. (See the appendix for additional discussion).

Antibody Panels and Staining Procedures

“Standard” antibody panels (Table 2) were used for identifying leukocyte populations and assessing expression of select cell surface proteins associated with innate and adaptive immunity, antigen presentation and inflammation. Anti-CD45 (leukocyte common antigen) was included in all sample tubes to help differentiate leukocytes from debris and assist in identifying leukocyte populations. Major lineage markers such as CD16, CD14, HLA-DR, CD3, CD19 and CD56 were used to aid in isolating specific populations. Certain antibodies were included in multiple sample tubes to facilitate gating of specific populations. Incorporation of additional fluorochromes would be advantageous, allowing consolidation of tubes and conservation of cells. Though we have done this in some panels for specific protocols, it is often difficult to procure specific antibodies labeled with the desired fluorochrome.

Table 2

Antibody combinations for leukocyte surface proteins associated with adaptive and innate immune responses.

TubeFITCPEPerCP, PE-Cy5APCAPC - Cy7Description
1IgG2aIgG2aIgG2aIgG2aCD45Isotype control
2IgG1IgG1IgG1IgG1CD45Isotype control
3CD64CD16CD11bCD14CD45IgG, CR3 and LPS receptor, Innate Immunity
4Lin-1*CD86HLA-DRCD11cCD45Antigen Presentation, Adaptive immunity
5Lin-1*CD83HLA-DRCD1aCD45DC maturity/activation, Adaptive immunity
6CD80CD40HLA-DRCD206CD45Antigen Presentation, Antigen Capture, Innate & Adaptive immunity
7CD282 (TLR2)CD284 (TLR4)HLA-DRCD14CD45Toll-like receptors, Innate immunity
8CD23 (FcεRII)FcεRIαCD19CD14CD45IgE receptor, Adaptive & innate immunity
9CD4CD8CD3CD25CD45Lymphocyte Populations
10CD19CD16/56CD3CD69CD45Lymphocyte Populations
*Lineage Cocktail-1 (BD Biosciences) – a mixture of anti-CD14, 16, 19, 20, 3, and 56.

CD45 is included in all tubes for leukocyte identification and gating. HLA-DR and CD14 are included in multiple tubes to facilitate differentiation and gating of specific populations.

Specific staining procedures have been described elsewhere (Alexis et al. 2000) and in the Appendix. Following staining, cells were fixed in 1% paraformaldehyde in Dulbecco’s phosphate-buffered saline (DPPS) and FCM performed within 24 hours. Stained samples should be refrigerated at 4°C until acquired. Sources for specific antibodies are listed in the appendix (Table 4). Specific antibodies were titrated to determine an appropriate amount for staining. We found it necessary to titrate and dilute isotype controls up to 1:5 (or more), especially for IgG2 isotypes since non-specific binding resulted in fluorescent intensity of the isotype control exceeding that of certain specific antibodies. Though fluorescence minus one (FMO) (Tung et al. 2004) controls would be preferable to isotype controls for determining background fluorescence, it was not feasible since it would require an excessive amount of cells to be performed properly. (See the appendix for additional discussion of isotype controls.)

Table 4

List of antibodies and cellular expression of target molecules.*

AntibodyCloneFluorochromeSourceCellular ExpressionFunction
CD1aBL6APCCoulterMono, Mac, DC, act. T & B cellsPresentation Of Non-Peptide Ag, Cell Mediated Immunity
CD313B8.2PerCPBDPan T cell MarkerComponent of T Cell Rcpt. Complex, T Cell Activation
CD4FICTCoulterT-helper cellsCo-Receptor For MHC-II Restricted T Cell Activation
CD8B9.11PECoulterCytotoxic T-cellsCo-Receptor For MHC-I Restricted T Cell Activation
CD11b/CR3ICRF44PE-CY5BDGran, Mac, MonoPhagocytosis, Cell Adhesion
CD11c/CR4B-ly6APCBDMac, Mono, GranPhagocytosis, Cell Adhesion
CD14RMO52APCCoulterMono, Mac, GranLipopolysaccharide (LPS) Receptor
CD16/FcγRIII3G8PECoulterPMN, Mac, NKPhagocytosis, Endocytosis
CD19J3-119FITCCoulterB cellsB Cell Marker, Component of B Cell Receptor Complex
CD23/FcεRII9P25FITCCoulterMono, Mac, B cellsLow Affinity IgE Receptor, Negative Feedback on IgE Production
CD25B1.49.9APCCoulterCD4(+) T-reg, Activated T&B cellsComponent of IL-2 Receptor
CD40MAB89PECoulterB cells, Mono, Mac, DC,B Cell Co-Stimulatory Molecule, Antigen Presentation, B Cell Maturation & Survival
CD452D1 N901APC-Cy7BDPan Leukocyte MarkerLeukocyte Common Antigen, B & T Cell Activation & Survival
CD56(NHK-1)FITCCoulterNK cells, NKT cellsNK Cell Marker, Cell Adhesion
CD64/FcγRI22FITCCoulterMac, Mono,Phagocytosis, Antigen Capture, Antibody Dependent Cellular Cytotoxicity
CD69FN50APCBDActivated T&B cells, PMNs & EOST Cell Activation, Activation Marker
CD80/B7.1MAB104FITCCoulterMac, Mono, DC,Co-Stimulatory Molecule For Antigen Presentation, T Cell Activation
CD83HB15aPECoulterMac, Mono, DCCo-Stimulatory Molecule For Antigen Presentation?
CD86/B7.2HA5.2B7PECoulterMac, Mono, DC, B cellsCo-Stimulatory Molecule For Antigen Presentation, T Cell Activation
CD206/Mannose Rcpt19.2APCBDMacrophagesC-Type Lectin Receptor, Phagocytosis, Antigen Capture
CD282/TLR2TL2.1FITCe-BioscMono, Mac, GranPathogen Associated Molecular Pattern (PAMP) Receptor
CD284/TLR4HTA125PEe-BioscMono, Mac, GranPAMP (LPS) Receptor
FcεRIαAER-37 (CRA1)PEe-BioscMono, Mac, basophils, mast cells,High-Affinity IgE Receptor, Type I Hypersensitivity
HLA-DRL243 (G46-6)PerCPBDMono, Mac, DC, B cellsMHC-II, Antigen Presentation
Lineage CocktailMultiple‡FITCBDNon-DC leukocytesLeukocyte Lineage Marker Cocktail
*There may be significant differential expression of these markers and some leukocytes not listed may express markers only at very low levels or following cell activation.
Coulter = Beckman Coulter, Inc., Brea, CA; BD = BD Biosciences, San Jose, CA ; e-Biosc = eBiosciences, San Diego, CA; Mac = macrophage; Mono = monocyte; Gran = granulocyte; DC = dendritic cells.
Clones used in Lineage cocktail: NCAM16.2, M[var phi]P9, L27, SJ25C1, 3G8, SK7.

Determination of Background Fluorescence Using Isotype Controls

Mean fluorescence intensity (MFI) of isotype controls (Tubes 1 and 2, Table 2) for specific populations was subtracted from the MFI measured for specific markers to control for background auto-fluorescence and non-specific fluorescence. For individual populations, background fluorescence was determined simultaneously for all fluorescence channels by creating a single histogram and gating 99% of the population (95% at the minimum) in only one fluorochrome channel, usually FITC. As illustrated for the monocyte population (Table 3), background MFIs for the various fluorochromes were relatively constant regardless of which channel was used for gating. This is also true for the other leukocyte populations and avoids the creation of a very large number of histogram gates.

Table 3

Determination of Background Fluorescence Using Isotype Controls.

PopulationGate Channel% GatedBackground MFI*
FITCPEPE-Cy5APC
MonocytesFITC99.0880295447717
PE99.0879283437718
PE-Cy599.0884286428714
APC99.9874287440709
NeutrophilsFITC99.0311170425734
MacrophagesFITC99.1726737044529776
LymphocytesFITC97.860240286532
*mean fluorescent intensity

Instrument Setup

We used a BD LSR-II digital flow cytometer equipped with 405nm solid state, 488nm argon-ion, and 633nm Helium-Neon lasers, appropriate filters and capability for cross-laser compensation (BD Biosciences Immunocytometry Systems, San Jose, CA). Our “standard” panel uses 5 colors exciting off the 488 and 633 lasers; however, additional fluorochromes, including those exciting off the 405 laser, are used in some studies. Compensations for spillover and spectral overlap were established for a particular set of instrument settings using BD CompBeads (BD Biosciences, cat 552843) and an automated compensation algorithm in BD FACSDiva 6.1 software (BD Biosciences).

The high auto-fluorescence of sputum macrophages, relative to lymphocytes, requires a compromise to allow application of a single set of PMT voltages for all populations. Our approach has been to optimize instrument settings for monocytes. (see Appendix)

STATISTICS

Statistical evaluation was performed using GraphPad Prism 5.03 software. Non-parametric statistics (Wilcoxon Signed Rank test or Mann-Whitney test) were used to compare groups. Spearman’s correlation was used to test correlation. Least-squares goodness of fit was used to calculate linear regression.

RESULTS

Identification of leukocytes in sputum samples

Figures 1 through through55 represent typical moderately contaminated sputum samples from healthy and mild asthmatic volunteer subjects and demonstrate a typical level of contaminating debris, moderate to high proportion of squamous epithelial cells and small proportion of EOS.

An external file that holds a picture, illustration, etc.
Object name is nihms317901f1.jpg
General Gating Strategies for Sputum Leukocyte Identification

(A) Relatively uncontaminated sputum sample. (B) A typical “dirty” raw sputum sample from a mild asthmatic with extensive contamination. Density plots (C – D) more clearly demonstrate location of neutrophils. (C) CD45(+) material including low FSC debris (D) due to non-specific staining or auto-fluorescence. (E) Viable cells were selected (P2) based on negative PI staining. (F) The majority of low FSC material was PI(+) non-viable cells and debris. (G) Cleaned CD45(+) cells showing general locations of cell populations. (H) Squamous epithelial cells are differentiated from macrophages as the CD206 negative population.

SSC = Side Scatter, FSC = Forward Scatter, Mono = monocyte,, PMN = neutrophil, Lymph = lymphocyte

An external file that holds a picture, illustration, etc.
Object name is nihms317901f5.jpg
Gating Strategies for Isotype Control Tubes

(A) Gating based on auto-fluorescence (FITC) vs. SSC better separates granulocytes from macrophages than does CD45 (see 1G). (B) EOS gated in the granulocyte population based on CD45 vs. SSC. (C) Gating of monocytes and macrophages (non-granulocytes). (D) Locations of EOS and DCs based on tube 4. Background fluorescence of monocytes is used as a surrogate for DCs.

SSC = Side Scatter, FSC = Forward Scatter, Mono = monocyte, Mac = macrophage. DC = dendritic cell, EOS = eosinophil, PMN = neutrophil

The levels of contamination in dot plots of a relatively uncontaminated and a more typical moderately “dirty” (i.e. contaminated) sputum sample are demonstrated in Figure 1A and B, respectively. Due to the broad range of cell size and granularity, forward and side scatter (FSC and SSC, respectively) was best displayed using a log scale.

In the majority of samples, leukocytes could not be reliably identified using light scatter properties alone, because they are intermingled with contaminating epithelial cells, detritus from dead cells, yeast, bacteria, micro-particles and exogenous inhaled particles. It was therefore essential to include anti-CD45 (leukocyte common antigen) in the antibody cocktail to facilitate leukocyte identification. Even though “leukocytes,” expressing CD45 could be segregated from the whole sample (Fig 1C), it is clear that, based on FSC (Fig 1D), a large proportion of the CD45(+) material is much smaller (lower FSC) than expected for leukocytes. We postulated that this is likely due to specific or non-specific CD45 staining of fragmented cell membranes and cell detritus, or diffusion of labeled antibody through leaky membranes of apoptotic, pyknotic or degenerate cells. This is supported by the fact that the majority of this CD45(+)/FSClow material stains positive with propidium iodide (PI), a cell-impermeant stain for viability, as demonstrated in Fig1E. To address this artifact, it was essential to exclude this material based on FSC (Fig1F, High FSC gate), especially for very “dirty” samples. All subsequent gating was done using the “cleaned” CD45 (+) cells (Fig 1F).

Leukocyte populations are similarly positioned in both blood and sputum, based both on light scatter properties and CD45 expression. Neutrophils (polymorphonuclear cells, PMNs) are usually abundant in sputum samples even in healthy individuals and, as in blood, lymphocytes are the smallest and least granular cells in sputum. PMN and, when present, lymphocyte populations served as convenient “landmarks” for locating sputum leukocytes by FCM. The use of density plots (as opposed to dot plots) can facilitate identification of PMN in sputum samples Fig 1C – H). The simplest gating strategy for leukocyte sub-populations was based on SSC and differential CD45 expression (Fig 1G). However, as demonstrated in the following sections, more complex strategies were required to adequately discriminate overlapping populations and subpopulations, especially small or rare-event populations (e.g. EOS, DCs, and basophils) and detection of dimly expressed surface proteins.

When present in sufficient number, squamous epithelial cells appeared as a separate population with high forward and side scatter (Fig 1B & C) and might easily be mistaken for macrophages. However, in contrast to macrophages, this population does not express CD206 (macrophage mannose receptor, Fig 1H) or HLA-DR (not shown) and was absent from samples having minimal squamous cells based on examination of stained slides (not shown).

Specific Gating Strategies

We have found that analysis was facilitated if lymphocyte and granulocyte populations were identified and gated first. Lymphocytes could be identified based on SSC and CD45 expression alone (Fig 2A). Granulocytes were identified within the non-lymphocyte gate (P2) using specific markers (e.g., CD64, CD16, CD14 or HLA-DR) which allowed for their discrimination either through positive or negative selection. Remaining non-granulocytes comprised monocyte, macrophage and DC populations, which were further differentiated based on differential SSC properties, CD45 expression and specific surface markers. Depending upon the antibody panel, it was not always possible to differentiate small populations such as DCs or EOS; however, inclusion of additional population-specific antibodies could facilitate their identification (see below).

An external file that holds a picture, illustration, etc.
Object name is nihms317901f2.jpg
Specific Gating Strategies: (Tube 3, 7, 8) Assessment of Innate Immune Markers

(A) Sputum lymphocytes are gated separately from non-lymphoctes (P2). (B) Differential expression of CD64 and CD16 facilitates separation of granulocytes (P3) from non-granulocytes. (C) Eosinophils are differentiated from PMNs by differential expression of CD16 (tube3). Remaining non-granulocytes from P3 are recombined with “Not P3” to comprise the Mono/Mac/DC population. (D) Macrophages and Mono/DCs gated base on CD45 vs. SSC properties. (E) CD14/CD16 dim dendritic cells (tube 3) fall primarily within the Monocyte gate (D). (F and G) Granulocytes are HLA dim/neg cells in tube 7, while DCs are CD14 dim/HLA high cells within the monocyte gate. (H) Granulocytes are CD14 dim/FcεR1 dim cells in tube 8.

SSC = Side Scatter, Mono = monocyte, Mac = macrophage. DC = dendritic cell, EOS = eosinophil, PMN = neutrophil,

Though differential expression of so called “lineage-specific” markers greatly facilitates discrimination of leukocyte populations, some overlap in expression of these molecules on various populations does occur (e.g. CD23 and CD14 on activated PMNs) and may result in less than optimal separation. As a result, gating necessarily becomes relatively more subjective and may vary depending upon the bias of individual analysts. Though this is largely unavoidable, subjectivity and misidentification of populations can be minimized by gating strategies based on two differentially-expressed markers or sequential “Boolean” gating strategies using two or more markers. The use of relatively conservative gating (i.e. gating tightly around populations) in such situations will also help to minimize misidentification of populations.

Evaluation of Innate Immune Response Proteins (Table 2, Tubes 3, 7 & 8)

Lymphocytes could be gated confidently based on SSC and CD45 expression (Fig 2A): however, antibodies against specific cell surface proteins were utilized to differentiate overlapping populations of PMNs, EOS and mononuclear phagocytes. Differential expression of CD64 and CD16 (tube 3) permitted isolation of PMNs, EOS and DCs from remaining monocytes and macrophages (Fig 2B). Neutrophils and EOS were differentiated based on CD16 expression (Fig 2 C). Keep in mind that degranulated EOS will have lower SSC properties and may blend with the PMNs based on SSC. The remaining non-granulocytes (Fig 2C) were monocytes, macrophages and DCs, which were recombined with the “not-P3”population (Fig 2B) and further partitioned on the basis of light scatter and differential expression of CD45, CD16 and CD14 (Fig 2D & E). Dendritic cells (Fig 2E) were identified as CD14dim/CD16dim and fell primarily within the monocyte population. Granulocytes in tube 7 (Fig 2F) were identified as HLA-DRdimCD14dim/neg cells and DCs could be identified as CD14dim//HLA-DRhigh non-lymphocytic cells (Fig 2G). The proportions of DCs identified in tubes 3 and 7correlated well with the more definitive identification of DCs and their identity can be verified using the lineage cocktail (not shown) as in tubes 4 and 5 (described below).

The expression of CD14 on activated PMNs (tube 8) often prevented good separation of PMNs from mononuclear phagocytes based solely upon CD14 expression. Differential expression of CD14 in combination with either CD23 or FcεRIα (dim expression on granulocytes) was used to facilitate isolation of granulocytes in this tube (Fig 2H). Though some activated PMNs and EOS may express low to moderate levels of these surface proteins, they are expressed at relatively low levels compared to mononuclear phagocytes. Inclusion of anti-HLA-DR would allow better discrimination of mononuclear phagocytes and identification of DCs.

Evaluation of Adaptive Immune Response Proteins (Table 2, Tubes 4, 5 and 6)

Dendritic cells were specifically identified (tubes 4 and 5) within the entire CD45(+) leukocyte population as Lineagedim/negative/HLA-DRhigh cells (Fig 3A). The lineage cocktail (Table 2) allowed differentiation of DCs from monocytes, macrophages, granulocytes, T-cells, B-cells and NK cells. Similar to blood, sputum DCs comprised a very small proportion (0.5 – 1.5%) of total sputum leukocytes and were dispersed mainly within the monocyte population (Fig 3B). The majority (≈ 90 to 95%) of sputum DCs were CD11c (+) myeloid DCs (Fig 3C), expressing high levels of CD86. Around 50% or more constitutively expressed CD83 while only a small proportion expressed CD1a (Fig 3D), suggesting that airway luminal DCs are relatively mature and at least partially activated. Subsets of DCs (Fig 3E) expressed both TLR2 and TLR4 (tube 7).

An external file that holds a picture, illustration, etc.
Object name is nihms317901f3.jpg
Specific Gating Strategies: (Tube 4, 5 & 6) Assessment of Adaptive Immune Markers

(A) Dendritic cells are lineage-dim/HLA-DR high cells from CD45(+) population and are located primarily within the monocyte gate (B). DCs are primarily CD11c(+) myeloid DCs (C) which express high levels of CD86. (D) The majority of DCs expressed CD83 (tube 5), while relatively few expressed CD1a. (E) Subpopulations of DCs (tube 7) expressed TLR2 and TLR4. Granulocytes gated based on differential expression of CD86 and lineage cocktail (F) or HLA-DR vs. SSC (G). A small proportion of granulocytes may express low levels of CD80 (tube 6) (H).

SSC = Side Scatter, DC = dendritic cell, EOS = eosinophil, PMN = neutrophil

Granulocytes were differentiated within the remaining (non-DC) cells based on a combination of SSC properties, differential expression of lineage markers and the negative or dim expression of either HLA-DR or co-stimulatory molecule CD86 (Fig 3F) in tube 4 or CD83 in tube 5. EOS were differentiated from PMNs as lineage-dim granulocytes (Fig 3F), similar to Fig 2C. Though granulocytes can usually be differentiated from mononuclear phagocytes on the basis of HLA-DR alone (Fig 3G), the use of a combination of two differentially expressed markers is preferable when possible. In most cases, the separation of granulocytes was well-defined; however, some PMNs and EOS may express low levels of HLA-DR and/or co-stimulatory molecules (CD80 and CD86), sometimes making the distinction between the populations less discrete. Mononuclear phagocytes were further dissected via SSC and differential expression of CD45 (see Fig 2D).

Differential expression of CD206 and HLA-DR (tube 6) allowed discrimination of granulocytes from macrophages (see Fig 1H). We found that CD80 was occasionally expressed at moderate levels on a small proportion of sputum granulocytes (Fig 3H). Though HLA-DR (+) lymphocytes are, for the most part, CD40 (+) B-cells (see Fig 4D) and also express some level of CD86, some activated T-cells may also express these proteins. Inclusion of additional antibodies to specifically identify B cells (tubes 4 and 5) and either lineage cocktail or CD14 to allow identification of DCs (tube 6) would be advantageous.

An external file that holds a picture, illustration, etc.
Object name is nihms317901f4.jpg
Sputum Lymphocyte Subpopulations

(A) CD3(+) T cells are comprised predominantly of CD4(+) T-helper cells. (B) NK cells and B cells comprise small and variable proportions of the lymphocyte population. (C) CD3(+) NK-T cells comprise the majority of sputum NK cells. (D) Sputum B cells express high levels of CD40. (E) Identification of NK cells (tube 3)

SSC = Side Scatter, FSC = Forward Scatter, NK = natural killer, T-ctl = cytotoxic T cell

Sputum Lymphocytes

Lymphocytes (tubes 9 and 10, Table 2) comprise a small and quite variable proportion of sputum leukocytes which is influenced by various factors including the health status of the individual and inflammatory processes in the bronchial airways. In this cohort of healthy,mild asthmatic and atopic individuals, the majority (≈ 80 to 90%) of sputum lymphocytes were CD3 (+) T cells, predominantly (≈ 60 to 65%) CD4 (+) T-helper cells (Fig 4A). B cells and natural killer (NK) cells comprised small and highly variable proportions (Fig 4B) of the lymphocytes. The majority of CD16/56 (+) NK cells were CD3 (+) NKT cells (Fig 4C). B cells (tube 6) were identified as HLA-DRhigh/CD40 high lymphocytes (Fig 4D). In the absence of CD56, NK cells could be tentatively identified as CD14dim/CD11bhigh/CD16high cells (tube 3) within the lymphocyte population (Fig 4E).

Gating for Isotype Controls

Measurement of background auto-fluorescence and non-specific staining was accomplished using isotype controls (Tubes 1 & 2 – Table 2). Lymphocytes could be gated easily since they do not overlap significantly with other populations. Simple gating of major populations based on SSC and CD45 expression may be adequate (see Fig 1G) for isotype controls; however, gating based on auto-fluorescence (FITC channel) and SSC (Fig 5A) usually provided better discrimination of overlapping granulocyte and macrophage populations. EOS overlap with both PMN and macrophage populations, while DCs are dispersed within the monocyte population. If present in sufficient numbers, the EOS gate could be based on the segregated granulocyte population (Fig 5B). After isolating granulocytes, the remaining monocytes and macrophages could be gated based on differential CD45 and SSC (Fig 5C) Regardless of which approach was used (FITC autofluorescence or CD45 vs. SSC), small populations such as EOS, DCs and basophils (extremely rare) were difficult (or impossible) to isolate without specific antibodies. Figure 5D shows the locations of the EOS and DC populations as determined by specific gating methods using tube 4 (see Fig 3 A and F). A simplistic alternative solution was to “non-specifically” gate the locations where these small populations are expected to exist or to substitute background auto-fluorescence values of their parent populations (i.e. monocytes for DCs, lymphocytes for basophils and PMNs for EOS). This is the approach we have taken for determining background fluorescence for DCs.

Specific Examples for Evaluating Airways Disease and Response to Inhaled Pollutants (Ozone)

The gating strategies and antibody panels described above are useful for monitoring expression of cell surface proteins or quantification and characterization of rare-event populations such as dendritic cells. The usefulness of these strategies for evaluating airways inflammation associated with asthma or exposure to environmental air pollution is demonstrated in Figure 6. As an example, using FCM, a slight yet significant (p<0.01) increase in the proportion of sputum dendritic cells was demonstrated (Fig 6A) in allergic individuals sensitive to dust mites (N=29; 19 mild allergic asthmatics and 10 allergic non-asthmatics) compared to healthy individuals (N=44). The value of flow cytometry in evaluating the effects of air pollutant exposure is exemplified by changes in surface expression of CD14 and HLA-DR on sputum monocytes (Fig 6B) from healthy volunteers 4 hours following a 2-hour inhalation exposure to 0.4 ppm ozone. (Hernandez et al. 2010).

An external file that holds a picture, illustration, etc.
Object name is nihms317901f6.jpg
Examples of Data Derived via Flow Cytometry of Sputum

(A) Proportions of sputum dendritic cells was significantly higher in dust-mite sensitive allergic individuals (N=29) compared to healthy non-allergic volunteers (N=44). (B) CD14 and HLA-DR expression was significantly higher on sputum monocytes from healthy volunteers 4hours after a 2-hr inhalation exposure to 0.4 ppm ozone (adapted from (Hernandez et al. 2010). (C) There was a significant (p=0.0001) correlation between %PMNs derived by FCM vs. stained slides (N=45). Filled circles = high viability (>70%) and low squamous cells (<35%). Open circles = low viability or high squamous cells, (D) Lymphocyte proportions tended to be higher based on FCM vs. stained slides.

Flow Cytometry-based Differential Cell Counts of Sputum Samples

Proportions of the various sputum leukocyte populations, as estimated by FCM, were similar but not identical to those determined by light microscopy. Differences between the two methods are variable and are affected by multiple factors, including the level of contamination of the sputum sample, cell viability, squamous cell count, and quality of the stained slide. For all major populations, as demonstrated for PMNs in Figure 6, there is generally greater correlation between methods when the sample is minimally contaminated and slide quality is high, as assessed by quality of staining and density of cells on the slide. Based on cell counts derived from tube 4, we found a significant correlation (p < 0.0001, Spearman’s r = 0.8233) between % PMN determined by FCM vs. microscopy (N=45 paired observations). The linear regression line (R2= 0.8856) and 95% confidence interval are shown (Fig 6C) for a subset of samples (N=30, filled circles) having cell viability ≥ 70% and squamous epithelial cell count ≤ 30% (based on differential counts from stained slides). Remaining samples (N=15, open circles) with higher squamous cell counts and lower viability are also plotted. When all 45 samples are included in the regression, R2 drops to 0.6658. Similar correlations were found for macrophages (not shown). Lymphocyte proportions, as determined by FCM, tended to be higher than those determined by microscopy (Fig 6D), as were those of EOS (not shown), both of which are present in relatively low proportions compared to PMNs and macrophages.

DISCUSSION

Inherent difficulties and artifacts peculiar to sputum render FCM challenging and necessitate specific gating strategies for both major and rare leukocyte populations such as DCs. Specifically, these include the presence of mucus, squamous and bronchial epithelial cells, dead and degenerate cells, cell detritus, microparticles and exogenous contaminants such as bacteria, yeast, pollen and inhaled dust particles. In addition, the presence of macrophages with inherently high auto-fluorescence necessitates a level of compromise when optimizing instrument settings. Unlike blood, overlapping major sputum leukocyte populations are not readily discernable and are further obscured by contaminating epithelial cells and debris which prevents efficient resolution of leukocyte populations by light scatter properties alone. Here we have outlined methods for overcoming these difficulties, including specific gating strategies for differentiating leukocytes from debris and identification of leukocyte subpopulations using a combination of light scatter properties and differential expression of CD45 and lineage-specific cell surface proteins. In addition, we have provided an approach for evaluating expression of cell surface proteins that are associated with both innate and adaptive immunity.

Samples with low viability are poorly suited for FCM due to excessive contamination with cellular debris, increased non-specific staining and increased auto-fluorescence. Our experience suggests that seventy percent viability or higher (50% at minimum) is desirable for FCM of sputum. Damaged cells may also generate staining artifacts in that labeled antibody may bind, specifically or non-specifically, to fragmented cell membranes and cell debris, or diffuse through leaky cell membranes of intact dead cells. This may allow inclusion of material with low FSC within the CD45(+) gate, thereby producing staining artifact (see Fig1C – F) and erroneous gating. Thus, a combination of both FSC and positive CD45 expression are required since, individually, neither adequately discriminate leukocytes from contaminants. While the use of isotype control antibody (and perhaps also unlabeled blocking antibody) may help to control for this artifact, the potential for erroneous results remains high when cell viability is low. It might still be possible to gain reliable data from samples with poor viability by segregating them using a viability marker such as propidium iodide (Fig 1E), 7-aminoactinomycin D, Calcein or other dyes. (Barry et al. 2002) Some newer products such as the “fixable” viability stains offered by Invitrogen (LIVE/DEAD® Fixable Dead Cell Stain Kit) may allow routine staining for both viability and surface markers followed by fixation and subsequent acquisition on the flow cytometer the next day. Though we have not yet tried the fixable dyes, they may work well for analysis of sputum samples.

Overlapping expression of various “lineage-specific” proteins on different cell populations may lead to a level of subjectivity in gating populations according to an individual’s own style or bias. Although this is unavoidable, subjectivity and bias can be minimized and objectivity increased by including antibodies to more than one differentially expressed marker instead of relying on a single marker. For example, in tube 3, the combination of CD64 and CD16 provides better separation of granulocytes than does CD16, CD14 or CD64 alone. Sequential “Boolean” gating may also be helpful in this respect. Over time, we continue to discover improvements which could be made in our antibody panels and have offered several suggestions as to how inclusion of certain alternate or additional antibodies in a specific tube might provide additional benefit (e.g. the addition of HLA-DR in tube 8). We have presented one approach to gating using tube-specific antibody panels; however, individual investigators can tailor panel configurations to suit their own research needs.

We emphasize that induction/collection and subsequent processing may significantly impact the quality of the sputum sample and subsequent analysis by FCM. Collection and processing of samples should be performed by well-trained and experienced personnel to maximize cell yield and viability and to minimize squamous cell contamination. Careful coaching of the subject/patient during expectoration can significantly reduce squamous cell contamination. While sample filtration will remove some proportion of the squamous cells, it cannot replace good induction technique. We have found that selectively extracting cell-rich mucus “plugs,” as opposed to processing the entire sputum sample, recovers the vast majority of the sputum leukocytes, reduces squamous cell contamination and saliva (Pizzichini et al. 1996) and greatly reduces the total volume of the sample to be processed, thereby reducing the dilution effect on fluid phase components of sputum.

Timely sample acquisition following antibody staining (ideally the same day) is desirable to maximize sample quality and minimize gating difficulties due primarily to paraformaldehyde-induced signal decay and increases in auto-fluorescence, both of which may occur over a relatively short period of time (24 – 48 hours). (Stewart et al. 2007) Although logistical issues may prevent same-day sample acquisition, we have found that acquisition within 24 hours of antibody staining does not compromise the integrity of the sample and subsequent data analysis.

Summary

The methods, antibody panels and gating strategies described here were developed over the course of ten years, evolving from two-color FCM, with gating based primarily on light scatter properties, to present day use of multi-color FCM and specific gating strategies which enable more accurate differentiation of sputum leukocytes from debris. Our experience has shown that care must be taken during sputum induction and processing to maximize the quality of the sample for FCM. Specific gating strategies based on a combination of light scatter properties, differential expression of CD45 and lineage-specific markers are essential for differentiating sputum leukocytes and their subpopulations from squamous epithelial cells and contaminating debris. Due to the significant overlap of major sputum leukocyte populations, it is necessary to include antibody against at least one cell surface marker (preferably two) which, either through positive or negative selection, will facilitate differentiation of granulocytes from mononuclear phagocytes. Once identified, additional FCM-based measurements of leukocyte surface receptors (immunophenotype) and function (phagocytosis, oxidative burst) can be performed. Formulation of antibody panels and gating strategies based on these principles greatly enhance the usefulness of multicolor FCM for the evaluation of sputum leukocytes.

Clinical Relevance

This research describes detailed methods to overcome difficulties associated with flow cytometry of sputum samples, which previously has been lacking in the literature. Flow cytometry of sputum samples can provide valuable information on inflammation and immunological response elements in the bronchial airways for both clinical diagnostic and research applications and can be a useful tool in inhalation toxicology for assessing health effects of inhaled environmental pollutants.

Acknowledgments

The authors wish to acknowledge and thank our highly skilled clinical and technical staff. These include Lynne Newlin-Clapp, Martha Almond, Carole Robinette, Margaret Herbst-Saunders, Aline Kala and Sally Ivins who perform sputum inductions, as well as Heather Wells, Fernando Dimeo, Danuta Sujkowski, Nolan Sweeney, and Katherine Mills who process the sputum samples and acquire data on the flow cytometer.

Funding: This research was funded in part by grants from The National Institutes of Health U19AI077437, R01-ES012706, RC1ES018417 and P01AT002620, as well as cooperative agreement CR 83346301 from the US Environmental Protection Agency. Although the research described in this article has been funded wholly or in part by the United States Environmental Protection Agency through cooperative agreement CR 83346301 with the Center for Environmental Medicine and Lung Biology at the University of North Carolina at Chapel Hill, it has not been subjected to the Agency’s required peer and policy review and therefore does not necessarily reflect the views of the Agency, and no official endorsement should be inferred.

Abbreviations

FCMflow cytometry
FSCforward scatter
SSCside Scatter
MFImean fluorescence intensity
DCdendritic cell
EOSeosinophil
PMNneutrophil
APCantigen presenting cell
TLRtoll-like receptor
LPSlipopolysaccharide
BALbronchoalveolar lavage
MHC-IImajor histocompatibility antigen class II

Appendix: Extended Methods and Comments

Sputum Sample Collection, Processing and Factors Affecting Sample Quality

Sputum induction and processing techniques vary and are described in detail elsewhere.(Kips et al. 1998b; Kips et al. 1998a; Ronchi et al. 2002; Efthimiadis et al. 2002b; Hargreave et al. 1998; Pizzichini et al. 1996; Spanevello et al. 1998) Methods used here are described in detail elsewhere (Alexis et al. 2000) and are based on the methods of Hargreave et al.(Hargreave et al. 1998) The quality of the sputum sample has major impact on the quality of the flow cytometric assessment and can be influenced both by the induction process and subsequent processing of the sample. Sputum samples should immediately be placed on ice and processed within 2 hours.(Efthimiadis et al. 2002a) Careful processing of the sample is crucial and it is important that both sputum induction and processing be performed by well-trained and experienced personnel to maximize cell yield and minimize squamous epithelial cell and salivary contamination. Excessive or prolonged exposure to dithiothrietol (DTT) during processing adversely affects both cell viability and surface marker expression (Efthimiadis et al. 1997; Loppow et al. 2000) and should be avoided. It is important to note that our laboratory employs the “plug selection” method where cell-enriched mucus “plugs” are manually selected from the surrounding clear saliva fluid and the total selected material is then processed, rather than the “whole sample” method, where the entire raw expectorated sample is processed. The main advantage of the plug-selection method is that it minimizes both squamous cell contamination and dilution from saliva. (Pizzichini et al. 1996; Spanevello et al. 1998; Alexis et al. 2006)

Construction of antibody panels

When constructing antibody panels for sputum analysis, it is essential to include anti-CD45 (leukocyte common antigen, pan leukocyte marker) in all sample tubes to help differentiate leukocytes from debris and facilitate identification of leukocyte populations. We take advantage of major lineage markers such as CD16, CD14, HLA-DR, CD3, CD19, CD56, as well as differential expression of other population specific markers as an aid in isolating specific populations and subpopulations. Certain specific antibodies are included in multiple sample tubes to aid in identifying specific populations for gating purposes. Using these principles, we have developed “standard” panels of fluorescent antibodies (See manuscript Table 2) for identifying leukocyte populations and assessing expression of select cell surface proteins associated with innate and adaptive immunity, antigen presentation and inflammation. Many of the innate immune proteins also interact with the adaptive immune system.

Cytometers equipped with multiple lasers are capable of simultaneously using many more than the five fluorochromes incorporated in our standard 5-color panel. It would be desirable to incorporate more fluorochromes in order to reduce duplication of certain markers, consolidate tubes and conserve cells. We have done this for certain of our more specific protocols, however it is often difficult (short of labeling antibodies in-house) to procure the desired antibody labeled with the necessary fluorochrome to accomplish this. Individual investigators can tailor antibody panels to incorporate additional fluorochromes and to fit their specific research objectives.

Staining Procedures

Cells should be labeled (stained) in a timely manner (as soon as possible) following sample processing and kept at 4 °C in the dark until acquired on the flow cytometer. Data acquisition should be done within 24 hours for best results.(Stewart et al. 2007) The specific antibody staining procedures have been described elsewhere.(Alexis et al. 2000) We routinely label 100 μl of cell suspension (1 × 106 cells/ml) based on total non-squamous nucleated cells (i.e. leukocytes and bronchial epithelial cells). Following staining, cells were fixed in 1% paraformaldehyde in Dulbecco’s phosphate-buffered saline (DPPS). Sources for specific antibodies used in these panels are listed in Table 4..

Specific antibodies were titrated to determine an appropriate amount for staining. The majority of the antibodies were still on the plateau of the titration curve when used at half the manufactures suggested amount (i.e. 10 μl vs. 20 μl). A few (e.g., CD16, CD3) required further dilution. Individual investigators should titrate their antibodies to determine appropriate concentrations for their specific applications. Isotype antibodies also were titrated; however, when we attempted to match protein concentrations and fluorochrome/protein concentrations, we found that the isotype controls had a much higher MFI signal than did certain of the specific antibodies, particularly for the IgG2 isotypes. We therefore found it necessary to dilute isotype control antibodies up to 1:5 (or more). The cause of this disparity is not entirely clear. (Hulspas et al. 2009) Some labs have advocated doing away with isotype controls for this and other reasons (Keeney et al. 1998) while others do not. (O’Gorman and Thomas1999). It may also be useful to employ nonspecific unlabeled blocking antibody to help minimize non-specific staining.

Alternatively, a “cleaner” method for determination of background fluorescence might be the use of “fluorescence minus one” (FMO) analysis (Tung et al. 2004), however, to be done properly, this requires large numbers of cells when multiple populations, multiple specific antibodies, multiple fluorochromes and two different isotypes (IgG1 and IgG2) are used. Since the number of leukocytes obtained from sputum samples is often limited, the FMO approach is usually not feasible for the typical sputum sample. Substitution of blood leukocytes for estimating background fluorescence (Dua et al. 2010) may be adequate for sputum lymphocytes, granulocytes and monocytes; however the absence of macrophages in blood is an obvious problem; although this might be overcome by “spiking” sputum isotype control tubes with blood leukocytes. In our experience, however, sputum monocytes tend to be more granular (higher SSC) with moderately higher autofluorescence (especially in the FITC channel) than peripheral blood monocytes.

Determination of Background Fluorescence Using Isotype Controls

Gating populations in the isotype control tubes is more complex for sputum than for blood since, unlike blood, major non-lymphocyte populations overlap and small or rare populations are imbedded within larger populations. More precise determination of background fluorescence for these small populations, requires inclusion of specific antibodies [e.g. CD9 or CD16 (EOS), CD203c (basophils), or CD14 (DCs)] in addition to CD45 in the isotype control tube.

Mean fluorescence intensity (MFI) of isotype controls (Tubes 1 and 2, Table 2) for specific populations was subtracted from the MFI measured for specific markers to control for background auto-fluorescence and non-specific fluorescence. For individual populations, background fluorescence was determined simultaneously for all fluorescence channels by creating a single histogram and gating 99% of the population (95% at the minimum) in only one fluorochrome channel, usually FITC. As illustrated for the monocyte population (Table 3), background MFIs for the various fluorochromes were relatively constant regardless of which channel was used for gating. This is also true for the other leukocyte populations. This approach avoided the creation of a very large number of histogram gates.

Instrumentation

The majority of our samples are acquired on a BD LSR-II digital flow cytometer equipped with 405nm solid state, 488nm argon-ion, and 633nm Helium-Neon lasers, appropriate filters and capability for cross-laser compensation (BD Biosciences Immunocytometry Systems, San Jose, CA). Compensation for spillover and spectral overlap are set for a particular set of instrument settings using BD CompBeads (BD Biosciences, cat 552843) and an automated compensation algorithm in BD FACSDiva 6.1 software (BD Biosciences). Once established, compensation values are not changed unless the established parameter settings are altered. Our “standard” panel uses 5 colors exciting off the 488 and 633 lasers; however, additional fluorochromes exciting off the 405 laser are used in some studies. This is determined by the availability of specific antibodies in a particular fluorochrome and the amount of sample available. While it is true that less sample is required when more fluorochromes are employed in a particular panel, the trade-off is that the complexity of instrument setup, compensation and data analysis also increases.

Instrument Setup, Data Acquisition and Special Considerations for Sputum Samples

The goal in establishing appropriate instrument settings is to establish the best signal to noise ratio which will provide a measurable signal, while minimizing the coefficient of variation of that signal. Optimal PMT voltages for samples containing mixed populations (e.g. blood) are usually established based on the population having the lowest auto-fluorescence (i.e. lymphocytes) and can be accomplished using cells or beads with low-range fluorescent intensity.(BD Application Note2000) The high auto-fluorescence of sputum macrophages, relative to lymphocytes, requires a compromise to allow application of a single set of PMT voltages for all populations. Our approach has been to optimize instrument settings for monocytes. The monocyte population, labeled only with CD45, is displayed in a separate histogram for each fluorochrome and voltages adjusted to place the entire monocyte population between 200 and 2000, which will place macrophages much higher on the log scale (between 5,000 and 10,000) and lymphocytes somewhat lower, perhaps even straddling zero. In the case of BD FACSDiva 6.1 software, bi-exponential plots allow the display of events with fluorescent intensity below zero. It is best, however, to use a setup optimized for lymphocytes if they are of primary interest.

Footnotes

DECLARATION OF INTERESTS

The authors report no declarations of competing interest.

References

  • Alexis N, Eldridge M, Reed W, Bromberg P, Peden DB. CD14-dependent airway neutrophil response to inhaled LPS: role of atopy. J Allergy Clin Immunol. 2001a;107:31–35. [Abstract] [Google Scholar]
  • Alexis N, Soukup J, Ghio A, Becker S. Sputum phagocytes from healthy individuals are functional and activated: a flow cytometric comparison with cells in bronchoalveolar lavage and peripheral blood. Clin Immunol. 2000;97:21–32. [Abstract] [Google Scholar]
  • Alexis NE, Hu SC, Zeman K, Alter T, Bennett WD. Induced sputum derives from the central airways: confirmation using a radiolabeled aerosol bolus delivery technique. Am J Respir Crit Care Med. 2001b;164:1964–1970. [Abstract] [Google Scholar]
  • Alexis NE, Lay JC, Zeman K, Bennett WE, Peden DB, Soukup JM, Devlin RB, Becker S. Biological material on inhaled coarse fraction particulate matter activates airway phagocytes in vivo in healthy volunteers. J Allergy Clin Immunol. 2006;117:1396–1403. [Abstract] [Google Scholar]
  • Alexis NE, Zhou H, Lay JC, Harris B, Hernandez ML, Lu TS, Bromberg PA, Diaz-Sanchez D, Devlin RB, Kleeberger SR, Peden DB. The glutathione-S-transferase Mu 1 null genotype modulates ozone-induced airway inflammation in human subjects. J Allergy Clin Immunol. 2009;124:1222–1228. [Europe PMC free article] [Abstract] [Google Scholar]
  • Antoniou KM, Alexandrakis M, Tzanakis N, Tsiligianni I, Tzortzaki EG, Siafakas NM, Bouros DE. Induced sputum versus bronchoalveolar lavage fluid in the evaluation of patients with idiopathic pulmonary fibrosis. Respiration. 2005;72:32–38. [Abstract] [Google Scholar]
  • Balbi B, Pignatti P, Corradi M, Baiardi P, Bianchi L, Brunetti G, Radaeli A, Moscato G, Mutti A, Spanevello A, Malerba M. Bronchoalveolar lavage, sputum and exhaled clinically relevant inflammatory markers: values in healthy adults. Eur Respir J. 2007;30:769–781. [Abstract] [Google Scholar]
  • Barry SM, Condez A, Johnson MA, Janossy G. Determination of bronchoalveolar lavage leukocyte populations by flow cytometry in patients investigated for respiratory disease. Cytometry. 2002;50:291–297. [Abstract] [Google Scholar]
  • Barry SM, Janossy G. Optimal gating strategies for determining bronchoalveolar lavage CD4/CD8 lymphocyte ratios by flow cytometry. J Immunol Methods. 2004;285:15–23. [Abstract] [Google Scholar]
  • BD Application Note. Establishing Optimum Baseline PMT Gains to Maximize Resolution on BD Biosciences Digital Flow Cytometers. (2000). 23–8389–00.
  • Bienkowska-Haba M, Cembrzynska-Nowak M, Liebhart J, Dobek R, Liebhart E, Siemieniec I, Panaszek B, Obojski A, Malolepszy J. Comparison of leukocyte populations from bronchoalveolar lavage and induced sputum in the evaluation of cellular composition and nitric oxide production in patients with bronchial asthma. Arch Immunol Ther Exp (Warsz) 2002;50:75–82. [Abstract] [Google Scholar]
  • Dominguez OJ, Leon F, Martinez Alonso JC, Alonso LA, Roldan E, Robledo T, Mesa M, Bootello A, Martinez-Cocera C. Fluorocytometric analysis of induced sputum cells in an asthmatic population. J Investig Allergol Clin Immunol. 2004;14:108–113. [Abstract] [Google Scholar]
  • Dua B, Watson RM, Gauvreau GM, O’Byrne PM. Myeloid and plasmacytoid dendritic cells in induced sputum after allergen inhalation in subjects with asthma. J Allergy Clin Immunol 2010 [Abstract] [Google Scholar]
  • Efthimiadis A, Jayaram L, Weston S, Carruthers S, Hargreave FE. Induced sputum: time from expectoration to processing. Eur Respir J. 2002a;19:706–708. [Abstract] [Google Scholar]
  • Efthimiadis A, Pizzichini MM, Pizzichini E, Dolovich J, Hargreave FE. Induced sputum cell and fluid-phase indices of inflammation: comparison of treatment with dithiothreitol vs phosphate-buffered saline. Eur Respir J. 1997;10:1336–1340. [Abstract] [Google Scholar]
  • Efthimiadis A, Spanevello A, Hamid Q, Kelly MM, Linden M, Louis R, Pizzichini MM, Pizzichini E, Ronchi C, Van Overvel F, Djukanovic R. Methods of sputum processing for cell counts, immunocytochemistry and in situ hybridisation. Eur Respir J Suppl. 2002b;37:19s–23s. [Abstract] [Google Scholar]
  • Frankenberger M, Menzel M, Betz R, Kassner G, Weber N, Kohlhaufl M, Haussinger K, Ziegler-Heitbrock L. Characterization of a population of small macrophages in induced sputum of patients with chronic obstructive pulmonary disease and healthy volunteers. Clin Exp Immunol. 2004;138:507–516. [Abstract] [Google Scholar]
  • Grootendorst DC, Sont JK, Willems LN, Kluin-Nelemans JC, Van Krieken JH, Veselic-Charvat M, Sterk PJ. Comparison of inflammatory cell counts in asthma: induced sputum vs bronchoalveolar lavage and bronchial biopsies. Clin Exp Allergy. 1997;27:769–779. [Abstract] [Google Scholar]
  • Hargreave FE, Pizzichini E, Pizzichini M. Induced sputum examination. J Allergy Clin Immunol. 1998;101:569–570. [Abstract] [Google Scholar]
  • Hernandez ML, Harris B, Lay JC, Bromberg PA, Diaz-Sanchez D, Devlin RB, Kleeberger SR, Alexis NE, Peden DB. Comparative airway inflammatory response of normal volunteers to ozone and lipopolysaccharide challenge. Inhal Toxicol. 2010;22:648–656. [Europe PMC free article] [Abstract] [Google Scholar]
  • Holden NJ, Bedford PA, McCarthy NE, Marks NA, Ind PW, Jowsey IR, Basketter DA, Knight SC. Dendritic cells from control but not atopic donors respond to contact and respiratory sensitizer treatment in vitro with differential cytokine production and altered stimulatory capacity. Clin Exp Allergy. 2008;38:1148–1159. [Abstract] [Google Scholar]
  • Hulspas R, O’Gorman MR, Wood BL, Gratama JW, Sutherland DR. Considerations for the control of background fluorescence in clinical flow cytometry. Cytometry B Clin Cytom. 2009;76:355–364. [Abstract] [Google Scholar]
  • Jaksztat E, Holz O, Paasch K, Kelly MM, Hargreave FE, Cox G, Magnussen H, Jorres RA. Effect of freezing of sputum samples on flow cytometric analysis of lymphocyte subsets. Eur Respir J. 2004;24:309–312. [Abstract] [Google Scholar]
  • Janossy G, Barry SM, Breen RA, Hardy GA, Lipman M, Kern F. The role of flow cytometry in the interferon-gamma-based diagnosis of active tuberculosis and its coinfection with HIV-1--A technically oriented review. Cytometry B Clin Cytom. 2008;74(Suppl 1):S141–S151. [Abstract] [Google Scholar]
  • Keeney M, Gratama JW, Chin-Yee IH, Sutherland DR. Isotype controls in the analysis of lymphocytes and CD34+ stem and progenitor cells by flow cytometry--time to let go! Cytometry. 1998;34:280–283. [Abstract] [Google Scholar]
  • Kips JC, Fahy JV, Hargreave FE, Ind PW, in’t Veen JC. Methods for sputum induction and analysis of induced sputum: a method for assessing airway inflammation in asthma. Eur Respir J Suppl. 1998a;26:9S–12S. [Abstract] [Google Scholar]
  • Kips JC, Peleman RA, Pauwels RA. Methods of examining induced sputum: do differences matter? Eur Respir J. 1998b;11:529–533. [Abstract] [Google Scholar]
  • Lay JC, Alexis NE, Kleeberger SR, Roubey RA, Harris BD, Bromberg PA, Hazucha MJ, Devlin RB, Peden DB. Ozone enhances markers of innate immunity and antigen presentation on airway monocytes in healthy individuals. J Allergy Clin Immunol. 2007;120:719–722. [Abstract] [Google Scholar]
  • Leckie MJ, Jenkins GR, Khan J, Smith SJ, Walker C, Barnes PJ, Hansel TT. Sputum T lymphocytes in asthma, COPD and healthy subjects have the phenotype of activated intraepithelial T cells CD69+ CD103+ Thorax. 2003;58:23–29. [Europe PMC free article] [Abstract] [Google Scholar]
  • Loppow D, Bottcher M, Gercken G, Magnussen H, Jorres RA. Flow cytometric analysis of the effect of dithiothreitol on leukocyte surface markers. Eur Respir J. 2000;16:324–329. [Abstract] [Google Scholar]
  • McCarthy NE, Jones HA, Marks NA, Shiner RJ, Ind PW, Al-Hassi HO, English NR, Murray CM, Lambert JR, Knight SC, Stagg AJ. Inhaled allergen-driven CD1c up-regulation and enhanced antigen uptake by activated human respiratory-tract dendritic cells in atopic asthma. Clin Exp Allergy. 2007;37:72–82. [Abstract] [Google Scholar]
  • O’Gorman MR, Thomas J. Isotype controls--time to let go? Cytometry. 1999;38:78–80. [Abstract] [Google Scholar]
  • Pizzichini E, Pizzichini MM, Efthimiadis A, Hargreave FE, Dolovich J. Measurement of inflammatory indices in induced sputum: effects of selection of sputum to minimize salivary contamination. Eur Respir J. 1996;9:1174–1180. [Abstract] [Google Scholar]
  • Pizzichini E, Pizzichini MM, Kidney JC, Efthimiadis A, Hussack P, Popov T, Cox G, Dolovich J, O’Byrne P, Hargreave FE. Induced sputum, bronchoalveolar lavage and blood from mild asthmatics: inflammatory cells, lymphocyte subsets and soluble markers compared. Eur Respir J. 1998;11:828–834. [Abstract] [Google Scholar]
  • Ronchi MC, Galli G, Zonefrati R, Tanini A, Scano G, Duranti R. Sputum processing: a new method to improve cytospin quality. Clin Exp Allergy. 2002;32:674–680. [Abstract] [Google Scholar]
  • Spanevello A, Beghe B, Bianchi A, Migliori GB, Ambrosetti M, Neri M, Ind PW. Comparison of two methods of processing induced sputum: selected versus entire sputum. Am J Respir Crit Care Med. 1998;157:665–668. [Abstract] [Google Scholar]
  • Stewart JC, Villasmil ML, Frampton MW. Changes in fluorescence intensity of selected leukocyte surface markers following fixation. Cytometry A. 2007;71:379–385. [Abstract] [Google Scholar]
  • Tung JW, Parks DR, Moore WA, Herzenberg LA, Herzenberg LA. New approaches to fluorescence compensation and visualization of FACS data. Clin Immunol. 2004;110:277–283. [Abstract] [Google Scholar]

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.3109/08958378.2011.575568

Supporting
Mentioning
Contrasting
4
52
1

Article citations


Go to all (37) article citations

Funding 


Funders who supported this work.

NCCIH NIH HHS (2)

NIAID NIH HHS (3)

NIEHS NIH HHS (7)