We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
ReportsOpen Accesscc iconby iconnc iconnd icon

Single-cell RNA sequencing of bronchoscopy specimens: development of a rapid minimal handling protocol

    Firoozeh V Gerayeli

    Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada

    ,
    Stephen Milne

    Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada

    Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada

    Sydney Medical School, University of Sydney, Camperdown, New South Wales, Australia

    ,
    Chen Xi Yang

    Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada

    ,
    Xuan Li

    Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada

    ,
    Elizabeth Guinto

    Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada

    ,
    Julia Shun-Wei Yang

    Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada

    ,
    Chung Yan Cheung

    Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada

    ,
    Tara R Stach

    Biomedical Research Center, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada

    ,
    Tawimas Shaipanich

    Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada

    ,
    Janice M Leung

    Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada

    Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada

    &
    Don D Sin

    *Author for correspondence: Tel.: +1 604 806 8396;

    E-mail Address: don.sin@hli.ubc.ca

    Center for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada

    Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada

    Published Online:https://doi.org/10.2144/btn-2023-0017

    Abstract

    Single-cell RNA sequencing (scRNA-seq) is an important tool for understanding disease pathophysiology, including airway diseases. Currently, the majority of scRNA-seq studies in airway diseases have used invasive methods (airway biopsy, surgical resection), which carry inherent risks and thus present a major limitation to scRNA-seq investigation of airway pathobiology. Bronchial brushing, where the airway mucosa is sampled using a cytological brush, is a viable, less invasive method of obtaining airway cells for scRNA-seq. Here we describe the development of a rapid and minimal handling protocol for preparing single-cell suspensions from bronchial brush specimens for scRNA-seq. Our optimized protocol maximizes cell recovery and cell quality and facilitates large-scale profiling of the airway transcriptome at single-cell resolution.

    Tweetable abstract

    We present a protocol for preparing single-cell suspensions from bronchial brushes for single-cell RNA sequencing that is fast, involves minimal handling, produces high cell yield/quality and is easily scalable due to efficiency and simplicity.

    Graphical abstract

    The airway mucosa plays a critical role in maintaining local and systemic homeostasis. Its protective functional properties include intercellular epithelial junctions, mucociliary clearance, soluble intraluminal immune factors, active immune surveillance and initiation of innate and adaptive cellular immunity [1]. Profiling the transcriptome of the airway mucosa has revealed insights into normal airway physiology as well as the pathophysiology of numerous diseases, including asthma [2–4], chronic obstructive pulmonary disease [5–8] and lung cancer [9,10]. However, bulk transcriptomics (either microarray or RNA sequencing) is unlikely to have sufficient resolution to capture the complexity of the airway mucosal cellular landscape.

    Single-cell RNA sequencing (scRNA-seq) allows analysis of the transcriptome at single-cell resolution. However, the majority of scRNA-seq data published to date have been derived from invasive airway biopsy, surgical resection or postmortem specimens. Such specimens are difficult to obtain and highly invasive and carry inherent safety risks. This presents a limitation to future large-scale scRNA-seq investigation of the airways, particularly in participants with comorbidities who have a higher risk of complications.

    Bronchial brush (BB) sampling of the airway mucosa is a less invasive and relatively safe procedure performed in routine clinical practice. A cytological brush is inserted via a bronchoscope into the airways and gently scraped against the airway wall to capture cells. In clinical practice, the specimens are assessed by cytology, microbiology or nucleic acid tests. In research settings, BB specimens have been used for profiling the airway transcriptome [2,3,6–9] and microbiome [11]. More recently, a number of studies have used BB specimens for scRNA-seq analysis of the airways [12–17].

    Despite many years of experience with research bronchoscopies and airway transcriptomics using BB specimens, our early attempts at preparing BB specimens for scRNA-seq were hampered by long processing times, low cell recovery, poor cell viability and low RNA quality. A review of the literature revealed significant heterogeneity among the previously published protocols used for this type of specimen (Table 1). We therefore aimed to develop a novel protocol for processing BB specimens for scRNA-seq based on the following guiding principles: minimize the handling and manipulation of cells, create a single-cell suspension with negligible cell clumping or debris, maximize cell recovery and viability and minimize processing time. Here we provide a qualitative description of the development of our optimized protocol for research. The resulting optimized protocol has facilitated a rapid expansion of our program, with over 40 specimens processed in the past 20 months. Given the inherent variability when working with clinical samples and to emphasize that, subsequent to protocol refinement, our methodology has yielded promising results across diverse disease conditions, we have provided the coefficient of variation for all sequenced samples to date in Supplementary Figure 5.

    Table 1. Summary of published protocols for bronchial brush processing for single-cell RNA sequencing.
    AuthorsChua et al. [12]Deprez et al. [13]Jaeger et al. [14]Vieira Braga et al. [15]Xu et al. [16]Zuo et al. [17]
    Sample collection
    Primary aimInvestigate COVID-19 mechanismsInvestigate cell population distributions along the airwaysCompare airway basal cells between different interstitial lung diseasesInvestigate the cellular landscape of the airways in health and asthmaInvestigate viral entry genes in upper and lower airwaysInvestigate the small airway epithelium in smokers and nonsmokers
    ParticipantsN = 2 critical COVID-19 patientsN = 9 healthyN = 15 interstitial lung diseaseN = 3 healthyN = 17N = 6
    Site of brushingLower airways (unspecified)9th- to 12th-generation bronchiSubsegmental bronchi, right lower lobe (three per participant)Not reportedRight main bronchus10th- to 12th-generation bronchi
    Sample processing
    MediaWashing: DMEM/F12 (cold)
    Final: PBS
    Washing: HBSS + 1% BSA (cold)
    Final: HBSS + 0.05% BSA (cold)
    Initial: BEGM (37°C)
    Freezing: 20% DMSO + 80% FBS BEGM
    Washing: PBS + 10% FBS (4°C)
    Final: PBS + 0.04% BSA
    Digestion: HBSS
    Washing, final: PBS + 1% BSA
    Not reportedFirst wash: small airway growth basal medium
    Second wash, final: PBS + 0.01% BSA
    Cell dissociationReduction: 13 mM DTT (10 min, 37°C)
    Digestion: Accutase™ (10 min, 37°C)
    Neutralization: DMEM/F12 + 10% FBS
    Digestion: 10 mg/ml Bacillus licheniformis protease/HypoThermosol/EDTA (30–60 min, cold)
    Neutralization: HBSS + 2% BSA
    Digestion: 0.05% trypsin/EDTA (5 min, 37°C)
    Neutralization: HEPES + 15% FBS
    Digestion: 1 mg/ml collagenase D + 0.1 mg/ml DNase (1 h, 37°C)Digestion: 0.25% trypsin/EDTADigestion: 0.05% trypsin/EDTA (5 min)
    Neutralization: HEPES + 15% FBS
    Red blood cell removalRed blood cell lysis buffer (10 min, 25°C) if macroscopic blood in sample0.8% ammonium chloride (10 min, cold) if red cell count >50% of total cellsNoRed blood cell lysis bufferGYPA/CD235a+ staining followed by exclusion by FACSAmmonium–chloride–potassium (3 min)
    Dead cell removalNoNoNoNoLive cells isolated by FACS (Hoechst 33342 + propidium iodide - CD235a-)DAPI-stained cell sorting (Influx cell sorter)
    scRNA-seq
    scRNA-seq platform10x Genomics Chromium (kit v3.1)10x Genomics Chromium (kit v2)10x Genomics Chromium (kit v2)10x Genomics10x GenomicsDrop-seq using FlowJEM microfluidic device
    Sequencing platformIllumina NovaSeq 6000Illumina NextSeq 500/550Illumina HiSeq 4000Illumina HiSeq 4000Illumina NextSeq 500Illumina HiSeq 2500
    Total cells after data processing15,24618,64417,3396761207511,702
    Average number of cells per participant (per brush)7623 (7623)2071 (2071)1155 (385)2253 (count unknown)122 (122)1950 (count unknown; previous protocol from this laboratory states 14 brushes per participant)
    Quality metrics and comments made by study authors Low fraction of reads per cell noted (range: 50–74%), suggesting high ambient RNA; ambient RNA removal (SoupX software) resulted in many zero count cells   Authors commented on selective drop-out of certain epithelial cell subpopulations (e.g., ciliated cells) during processing; ambient RNA 5.4–6.2%

    †Calculated as total cells/(number of participants) * (number of brushes per participant).

    BEGM: Bronchial Epithelium Cell Growth Medium; BSA: Bovine serum albumin; DAPI: 4′,6-diamidino-2-phenylindole; DMEM: Dulbecco’s modified Eagle medium; DMSO, dimethyl sulfoxide; Drop-seq: Droplet sequencing; DTT: Dithiothreitol; FACS: Fluorescence-activated cell sorting; FBS: Fetal bovine serum; HBSS: Hanks' Balanced Salt Solution; HEPES: N-2-hydroxyethylpiperazine-N-2-ethane sulfonic acid; PBS: Phosphate-buffered saline; scRNA-seq: Single-cell RNA sequencing.

    Materials & methods

    Human research ethics

    The specimens used for this study were collected under protocols that were approved by the Human Research Ethics Board of the University of British Columbia/Providence Health Care (approval nos. H11-02713, H19-02222 and H21-02149). All study participants provided informed consent.

    BB protocol

    The details of our standardized research bronchoscopy protocol have been published previously [18]. After receipt of informed consent and under conscious sedation, a fiberoptic bronchoscope (Olympus Corporation, Tokyo, Japan) was passed through the mouth of participants and into the trachea. With the bronchoscope positioned in one of the subsegmental bronchi of the right or left upper lobe, a cytological brush was inserted through a bronchoscope channel into a sixth- to eighth-generation airway, from where BB samples were collected. The cytological brush was then withdrawn from the bronchoscope, and using a pair of stainless steel scissors, the brush was cut into a microcentrifuge tube containing 1000 μl medium and kept on ice until further processing. We evaluated the following media for sample quality (Table 2): Dulbecco's phosphate-buffered saline (DPBS) (Thermo Fisher Scientific, MA, USA), Roswell Park Memorial Institute 1640 (RPMI1640) (Corning, NY, USA) with and without fetal bovine serum (Thermo Fisher Scientific, MA, USA) and PneumaCult-Ex™ culture medium (STEMCELL Technologies, Vancouver, Canada).

    Table 2. Bronchial brush protocol optimization and outcomes.
    ParticipantsParticipant 001Participant 002Participant 003Participant 004Participant 005Participant 006Participant 007Participant 008Participant 009
    Participant characteristics66-year-old male with COPD75-year-old female with lung mass42-year-old female with asthma69-year-old male with no prior history of lung disease71-year-old male with lung mass60-year-old male with COPD32-year-old female with no prior history of lung disease recovered from COVID-1959-year-old female with COPD62-year-old male with COPD
    Sample preparation
    MediumDPBSDPBSDPBS + 10% FBSDPBS + 10% FBSDPBS + 10% FBSRPMI 1640PneumaCult-Ex™PneumaCult-Ex™PneumaCult-Ex™
    TemperatureRoom temperatureRoom temperatureRoom temperatureRoom temperatureRoom temperatureRoom temperatureSpecimens and reagents kept on ice; centrifuged at 4°CSpecimens and reagents kept on ice; centrifuged at 4°CSpecimens and reagents kept on ice; centrifuged at 4°C
    Cell dissociationAccutase™Accutase™Accutase™Accutase™ + plate shakerAccutase™ + plate shakerAccutase™ + plate shakerAccutase™ + plate shakerAccutase™ + gentle agitationAccutase™ + gentle agitation
    Protease incubation time, min1010101010101033
    Wash–centrifuge steps1 × 5-ml wash1 × 5-ml wash3 × 5-ml wash3 × 5-ml wash3 × 25-ml wash1 × 25-ml wash1 × 25-ml wash1 × 25-ml wash1 × 25-ml wash
    Filtration40-μm bucket strainer40-μm bucket strainer40-μm bucket strainer40-μm bucket strainer40-μm bucket strainer40-μm bucket strainer70-μm bucket strainer70- and 40-μm bucket strainers40-μm bucket and Flowmi™ strainers
    Final volume, μlUnknown150150Unknown30010050100100
    Final concentration (hemocytometer), cells/μlCell morphology was determined to be abnormal; no cell count was doneNo viable cells detected for cell countNo viable cells detected for cell count after all three washesNo viable cells detected for cell count after all three washesNo viable cells detected for cell count after all three washes500200333505
    Morphology commentsAbnormal cell morphology (very small, round) (Supplementary Figure 3)Dead/fragmented cells, residual debrisDead/fragmented cells, abnormal morphology, debris and cell clumps after second washDead/fragmented cells, abnormal morphology, debris and cell clumps after second washDead/fragmented cells, abnormal morphology, debris and cell clumps after second washAbnormal cell morphology (small, round)Abnormal cell morphology (small, round)Ciliated, other epithelial and immune cell morphologies observedCiliated, other epithelial and immune cell morphologies observed
    scRNA-seq
    scRNA-seq platformNot submittedNot submittedNot submittedNot submittedNot submitted10x Genomics Chromium (kit v3.1)10x Genomics Chromium (kit v3.1)10x Genomics Chromium (kit v3.1)10x Genomics Chromium (kit v3.1)
    RNA sequencingNot submittedNot submittedNot submittedNot submittedNot submittedIllumina NextSeqIllumina NextSeqIllumina NextSeqIllumina NextSeq
    Number of cells calledNot submittedNot submittedNot submittedNot submittedNot submitted57260510171102
    Data quality commentsNot submittedNot submittedNot submittedNot submittedNot submittedRNA fragmentation/high ambient RNA; low fraction of reads in cellsRNA fragmentation/high ambient RNA; low fraction of reads in cellsLow fraction of reads in cells (ideal >70%)Low fraction of reads in cells (ideal >70%)
    Cell Ranger comment (fraction of reads in cells), %Not submittedNot submittedNot submittedNot submittedNot submitted39.420.167.866.4
    Cell Ranger comment (reads mapped confidently to genome), %Not submittedNot submittedNot submittedNot submittedNot submitted54.774.478.974.7
    SoupX global ρ estimationNot submittedNot submittedNot submittedNot submittedNot submittedHigh contamination fraction; no plausible marker genes found0.070.010.01

    COPD: Chronic obstructive pulmonary disease; DPBS: Dulbecco's phosphate-buffered saline; FBS: Fetal bovine serum; RPMI 1640: Roswell Park Memorial Institute 1640; scRNA-seq: Single-cell RNA sequencing.

    For enzymatic digestion of intercellular junctions and extracellular matrix, approximately 1000 μl of Accutase™ (Thermo Fisher Scientific) was added directly to brush specimens in the medium followed by gentle agitation. The digestion time was titrated from 3 to 10 min, and the enzyme activity was quenched by directly transferring the combined Accutase™/medium into a separate tube containing either 2 ml (‘small-volume protocol’) or 25 ml (‘large-volume protocol’) of medium.

    To wash the cells, the cell suspension was passed through a 70-μm cell strainer (VWR, PA, USA) to remove large debris and cell clumps and centrifuged at 400 × g for 10 min at 4°C, after which the supernatant was discarded. Under the small-volume protocol, the cell pellet was resuspended in 2 ml medium and centrifugation was repeated as described, whereas the large-volume protocol included a single centrifugation step. The final cell pellet was resuspended in medium to a total volume of 100 μl, which was then filtered through either a 40-μm cell strainer (VWR) or a 40-μm Flowmi™ strainer (SP Bel-Art, PA, USA).

    Single-cell RNA sequencing

    Visual assessments and cell counts of the final cell suspensions were performed using a hemocytometer with trypan blue stain (Thermo Fisher Scientific). When necessary, the cell suspensions were diluted to a concentration of 1000 cells/μl in their respective media prior to single-cell sequencing. For further examination, aliquots of the cell suspensions were applied to histological slides by cytospin and stained using a modified Wright–Giemsa stain.

    Cell capture & library preparation

    Cell suspensions were transferred on ice to our sequencing facility (approximately 25 min away) and loaded onto the Chromium Controller or Chromium X using the Chromium Next GEM Single Cell 3′ Kit v3.1 and Chip G (10x Genomics, Inc., CA, USA). Samples were handled and libraries prepared according to the published Chromium Single Cell 3′ Reagent Kits User Guide [19]. The quality of the cDNA libraries and final DNA libraries was analyzed using the 2100 Bioanalyzer instrument with the High Sensitivity DNA Kit (Agilent Technologies, Inc., CA, USA).

    Sequencing

    Final libraries were sequenced on the NextSeq 2000 (Illumina, Inc., CA, USA) as recommended by 10x Genomics (read 1: 28 bp; read 2: 90 bp; i7 index: 10 bp; i5 index: 10 bp) at a loading concentration of 650 pM with a 2% PhiX spike-in (Illumina, Inc.). All sequencing was initially performed at a shallow depth of 20,000 reads/cell and assessed, and then further sequencing was performed to a depth of 60,000 reads/cell. Fastq files were generated with Cell Ranger v6.01 (10x Genomics) using the default options, and reads were aligned to the human reference genome (hg19).

    Bioinformatics

    A detailed description of the data processing is provided in Supplementary Figure 1. Analysis of scRNA-seq data can be challenging because of the presence of technical artifacts, such as ambient RNA. Such contamination would inadvertently impact the downstream analysis of the samples. The SoupX package (version 1.5.2) [20] in R was used prior to performing individual quality control tasks to correct for ambient RNA, whose expression is projected from the empty droplet pool. Following ambient RNA correction, using the Pegasus package (version 1.5.0) [21,22] in Python, we performed additional quality control steps, in which cells with high mitochondrial genes (>20%) and <200 genes per cell were filtered out. This step was followed by log normalization, batch effect correction using the harmony algorithm and doublet removal. We then performed principal component analysis prior to applying Uniform Manifold Approximation and Projection dimensionality reduction. Before performing any further downstream analysis, we annotated the data set using automatic annotation in the Pegasus package in conjunction with the markers noted in the literature.

    Results & discussion

    The central motivation for our work was to investigate the pathology of airway diseases using scRNA-seq. Bronchial brushing is an attractive method for sampling the lung because it is relatively noninvasive and safe and is routinely performed in clinical practice. A number of investigators have reported scRNA-seq using BB specimens [12–17]. However, a review of these published protocols revealed key differences in the way the specimens were processed (Table 1). Additionally, the number of cells recovered per participant was highly variable, and none of the published protocols were accompanied by detailed validation of their results. We therefore sought to develop our own protocol that maximized cell recovery and quality. The final BB protocol and, for the sake of completeness and in the interest of other investigators, the optimized protocol for preparing bronchoalveolar lavage specimens as well as a Uniform Manifold Approximation and Projection of one of the bronchoalveolar lavage samples we submitted for sequencing using this protocol are provided in Supplementary Figure 2.

    The choice of medium is important not only for maintaining cell viability but also for supporting single-cell suspension. The 10x Genomics standard protocol [23] recommends a calcium/magnesium-free balanced salt solution spiked with protein, which minimizes cell losses and aggregation. Indeed, the published BB protocols used either fetal bovine serum or bovine serum albumin (Table 1). However, we found that the use of these solutions for BB specimens was associated with poor cell viability and excessive clumping of cells, which led to low concentrations of viable cells at the end of processing. We found that the best medium was PneumaCult-Ex™, a serum- and bovine pituitary extract-free cell culture medium specifically formulated to support the growth of airway epithelial cells. We used this medium at all stages of processing BB specimens in the optimized protocol. To the best of our knowledge, this is the first time this medium has been used for scRNA-seq experiments.

    In order to generate a single-cell suspension from BB specimens, it was necessary to incubate the brushes in a protease solution. Enzymatic digestion of whole tissue and biopsy specimens is routinely used to break down extracellular matrix and create single-cell suspensions. Although BB specimens contain little extracellular matrix, the epithelial cells scraped from the airways were connected by intercellular junctions and therefore tended to remain in sheet-like clumps (Figure 1). The use of protease treatment of BB specimens was common to all previously published protocols, though most employed nonspecific proteases, such as trypsin (Table 1). We elected to use Accutase™, an enzyme mixture with proteolytic and collagenolytic properties, since it is associated with efficient cell dissociation without interrupting cell surface antigens or inducing cell cycle changes [24] and leads to improved viability when culturing fragile cell types [25]. Digestion for 10 min, as used by Chua et al. [12], resulted in widespread cell destruction and fragmentation, with occasional single cells that lacked the expected morphology of bronchial epithelial cells (Figure 2A & Supplementary Figures 3 & 4). After single-cell capture and cDNA library preparation, the Bioanalyzer indicated high levels of short sequences suggestive of either high ambient RNA or fragmented RNA in the specimens (Figure 2B). The processed data confirmed high levels of ambient RNA, a low number of cells (far below the expected number based on the sample input) and only three identifiable cell clusters (Figure 2C). Notably, the proportion of epithelial cells was very low (Figure 2D), suggesting that this cell type is particularly sensitive to the effects of prolonged protease activity. By contrast, Accutase™ digestion for 3 min produced a single-cell suspension with readily identifiable bronchial epithelial cells, including ciliated and goblet cells, as well as immune cells, such as monocytes, and the resulting cDNA Bioanalyzer output showed superior quality of RNA (Figure 3A). The processed scRNA-seq data demonstrated cell numbers closer to the expected count, with a diverse population of epithelial and immune cells consistent with the expected composition of the airway mucosa (Figure 3B & C). We therefore concluded that 3 min was the optimal duration of Accutase™ incubation.

    Figure 1. Bronchial brush specimen, untreated.

    Cells were retrieved from a single airway by bronchial brushing. The brush was immediately placed in PneumaCult-Ex™ medium, gently vortexed and centrifuged at 400 × g for 10 min at 4°C. The majority of cells were epithelial cells in large sheet-like clumps, with scattered immune cells and acellular debris. Modified Wright–Giemsa stain, ×40 magnification.

    Figure 2. Bronchial brush specimen of a 60-year-old male (Participant 006) with chronic obstructive pulmonary disease, 10-min Accutase™ digestion.

    A total of 1 ml Accutase™ was directly added to the brush in medium for 10 min on a plate shaker. Digestion was stopped by adding an excess of medium. The sample was then centrifuged at 400 × g for 10 min at 4°C. (A) Modified Wright–Giemsa stain, ×40 magnification, representative of observed widespread cell destruction and fragmentation and occasional small cell lacking known epithelial cell morphology. (B) Bioanalyzer output of the cDNA library following cell capture and reverse transcription PCR showing high number of short sequences suggestive of RNA fragmentation. (C) UMAP of single-cell RNA sequencing data showing three cell clusters. (D) Cell proportions from single-cell RNA sequencing, with a minority annotated as epithelial cells.

    UMAP: Uniform Manifold Approximation and Projection.

    Figure 3. Bronchial brush specimen of a 59-year-old female (Participant 008) with chronic obstructive pulmonary disease, 3-min Accutase™ digestion.

    A total of 1 ml Accutase™ was added directly to the brush in medium for 3 min with gentle agitation. Digestion was stopped by adding an excess of medium. The sample was then centrifuged at 400 × g for 10 min at 4°C. (A) Bioanalyzer output of the cDNA library following cell capture and reverse transcription PCR. (B) UMAP of single-cell RNA sequencing data showing multiple cell clusters of both epithelial and immune cell lineages. (C) Cell proportions from single-cell RNA sequencing.

    Anno: Annotation; FU: Fluorescent units; UMAP: Uniform Manifold Approximation and Projection.

    Washing with medium followed by centrifugation not only removes ambient (i.e., extracellular) RNA but also eliminates residual proteases from the cells. The previously published protocols for BB specimens as well as the 10x Genomics standard protocol [23] employed a minimum of two wash–centrifuge cycles (Table 1). However, we found that multiple wash cycles led to excessive cell loss. We therefore employed a single large-volume wash of 25 ml PneumaCult-Ex™ for BB specimens, which greatly improved the cell yield while minimizing ambient RNA. We also removed debris and cell clumps using mechanical filtration. The presence of cell clumps not only leads to multiplets in the sequencing output (i.e., more than one cell being captured in an oil droplet) but also interferes with the microfluidic technology and generation of the gel emulsion [23]. We found that a 40-μm cell strainer was adequate to remove large clumps and debris, such as mucus, as an initial step. However, the cell strainer required adequate rinsing with clean medium to minimize the loss of single cells captured by the strainer. Since cell retrieval from BB specimens was typically low, this use of extra medium further diluted our cell concentrations. In order to minimize the volume and therefore increase cell concentration, we used a 40-μm Flowmi™ cell strainer as the final filtration step. This filter attaches directly to the pipette tip and can be used to filter volumes as low as 50 μl with minimal volume loss. In our experience, we were able to filter volumes of 100–120 μl, with a typical volume loss of 20–40 μl.

    To optimize the BB specimen protocol, there were several other considerations. The 10x Genomics standard protocol [23] and some of the published protocols [16,17] employed a dead cell removal step (e.g., flow cytometry) in order to maximize the viable cell concentration. However, we elected to forgo this step for three reasons. First, our cell recovery was typically low, and dead cell removal would further reduce the final cell concentration. Second, we considered our cells fragile and needed to avoid further mechanical stress. Third, the quality of our cells was highly dependent on the time since collection, meaning delays related to this step would further compromise the overall quality of the cells. We also did not employ red blood cell lysis techniques since our bronchial epithelial cells were sensitive to the typical lysis reagents (e.g., ammonium chloride). Instead, we avoided using macroscopically contaminated specimens and removed any red blood cell ‘contaminants’ in silico using bioinformatics tools. We kept specimens and reagents at 4°C rather than at room temperature (as recommended by the 10x Genomics standard protocol) [23] because, in our experience, this minimized cell clumping. Finally, we used low retention materials (conical and microcentrifuge tubes) wherever possible to maximize cell recovery. With each of these measures, our BB specimen protocol produced high-quality single-cell suspensions with increased and reliable cell counts (Table 2). The processed sequencing data showed that all of the expected cell types in the airway epithelium were present (Figure 4). Demographic information pertaining to the data presented in Figure 4 can be found in Supplementary Figure 6.

    Figure 4. Combined single-cell RNA sequencing output using the optimized protocol.

    (A) UMAP of single-cell RNA sequencing data of five consecutive subjects, recruited after optimization of the protocol, showing various cell clusters. Post-quality control average cells per brush = 2349 ± 1680 (mean ± standard deviation). (B) Cell proportions from single-cell RNA sequencing, consisting of annotated epithelial and immune cells.

    Anno: Annotation; UMAP: Uniform Manifold Approximation and Projection.

    Limitations

    At the time of this protocol development, we faced multiple challenges, including operating in the midst of a once-in-a-generation pandemic, which severely limited our access to human samples; the scarcity of samples available for replication/validation since each participant was enrolled under a set approved protocol for specimen collection; the extremely poor quality of cells in the early stages, which limited our ability to provide even the most basic quantitative metrics (e.g., cell count and viability); and the prohibitively high cost of sequencing suboptimal specimens. These challenges contributed to some important limitations. First, the limited sample availability meant that we were unable to systematically vary the experiments one variable at a time. It also restricted our ability to obtain technical replicates for each of the experimental conditions. Taken together, we were not able to robustly assess the effects of varying each condition and instead relied on qualitative descriptions of single trials. We included quantitative measures such as cell counts and sequencing output wherever possible. We purposefully avoided the use of cultured cells to optimize the protocol since, in our experience, primary BB cells do not behave in the same way as cultured cells, especially with regard to their fragility. Second, the bronchoscopy samples were processed immediately following bronchoscopy, which kept the overall time from cell collection to capture short. Having the bronchoscopy suite and the processing lab in the same building might not be the case for all research centers, and as such our protocol might not be generalizable to laboratories with different setups. Nevertheless, we believe that overcoming such logistical difficulties to maximize efficiency and minimize processing time should be a guiding principle for all laboratories performing scRNA-seq of BBs regardless of the setup.

    Conclusion

    We developed a protocol for preparing BB and bronchoalveolar lavage specimens for scRNA-seq that maximizes cell viability and recovery by minimizing specimen handling and processing time. The use of this optimized protocol has allowed us to build an scRNA-seq data set from over 50,000 cells, representing the airways of 26 participants. This large data set will be used to explore the pathology of various airway diseases and how changes in the airways at single-cell resolution may relate to clinically important outcomes.

    Executive summary

    Background

    • Single-cell RNA sequencing (scRNA-seq) allows analysis of the transcriptome at single-cell resolution.

    • scRNA-seq reveals heterogeneity in the airway cellular landscape.

    • Currently, the majority of data published on scRNA-seq have been derived from surgical biopsies; surgical resection of tissues, including explants; or postmortem specimens.

    • Bronchial brush sampling of the airway mucosa may be a safer and less invasive alternative method for investigating airways.

    Materials & methods

    • We have developed a protocol for creating single-cell suspensions from bronchial brush samples with negligible cell clumping and debris while minimizing processing time.

    Results & discussion

    • Our protocol will facilitate and standardize large-scale scRNA-seq experiments, enabling new insights into the pathophysiology of airway diseases.

    Supplementary data

    To view the supplementary data that accompany this paper please visit the journal website at: www.future-science.com/doi/suppl/10.2144/btn-2023-0017

    Author contributions

    FV Gerayeli, S Milne, J Shun-Wei Yang and CY Cheung developed the laboratory protocols. FV Gerayeli, S Milne, E Guinto, TR Stach, T Shaipanich and JM Leung collected and processed the specimens. CX Yang developed the bioinformatics pipeline. FV Gerayeli, CX Yang and X Li analyzed the data. FV Gerayeli and S Milne wrote the first draft manuscript. DD Sin had full oversight of the research and has full access to the data. All authors reviewed, edited and approved the final manuscript.

    Financial disclosure

    The study was supported by grants from the Canadian Institutes of Health Research (GA4-177740 and PJT-178058), Genome British Columbia (SIP029), Michael Smith Health Research BC (RT-2022-2661) and Mitacs (IT13817 and IT28140). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Competing interests disclosure

    S Milne has received financial support from Chiesi Australia for work performed outside of the present study. DD Sin has received honoraria from GlaxoSmithKline, AstraZeneca and Boehringer Ingelheim for work performed outside of the present study. The authors have no competing interests or relevant affiliations with any organization or entity with an interest in or conflict with the subject matter or materials discussed in the manuscript.

    Writing disclosure

    No writing assistance was utilized in the production of this manuscript.

    Ethical conduct of research

    The authors state that they have obtained appropriate institutional review board approval for all human experimental investigations. In addition, informed consent has been obtained from the participants involved.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest

    References

    • 1. Frey A, Lunding LP, Ehlers JC, Weckmann M, Zissler UM, Wegmann M. More than just a barrier: the immune functions of the airway epithelium in asthma pathogenesis. Front. Immunol. 11, 761 (2020).
    • 2. Chun Y, Do A, Grishina G et al. Integrative study of the upper and lower airway microbiome and transcriptome in asthma. JCI Insight 5(5), e133707 (2020).
    • 3. Modena BD, Bleecker ER, Busse WW et al. Gene expression correlated with severe asthma characteristics reveals heterogeneous mechanisms of severe disease. Am. J. Respir. Crit. Care Med. 195(11), 1449–1463 (2017).
    • 4. Park HW, Weiss ST. Understanding the molecular mechanisms of asthma through transcriptomics. Allergy Asthma Immunol. Res. 12(3), 399–411 (2020).
    • 5. Chang WA, Tsai MJ, Jian SF, Sheu CC, Kuo PL. Systematic analysis of transcriptomic profiles of COPD airway epithelium using next-generation sequencing and bioinformatics. Int. J. Chron. Obstruct. Pulmon. Dis. 13, 2387–2398 (2018).
    • 6. Milne S, Li X, Yang CX et al. Inhaled corticosteroids downregulate SARS-CoV-2-related genes in COPD: results from a randomised controlled trial. Eur. Respir. J. 58(1), 2100130 (2021).
    • 7. Morrow JD, Chase RP, Parker MM et al. RNA-sequencing across three matched tissues reveals shared and tissue-specific gene expression and pathway signatures of COPD. Respir. Res. 20(1), 65 (2019).
    • 8. Steiling K, van den Berge M, Sebastiani P et al. Airway gene expression as a molecular phenotype of COPD. Proc. Am. Thorac. Soc. 8(2), 208 (2011).
    • 9. Beane J, Vick J, Schembri F et al. Characterizing the impact of smoking and lung cancer on the airway transcriptome using RNA-seq. Cancer Prev. Res. (Phila.) 4(6), 803–817 (2011).
    • 10. Boelens MC, van den Berg A, Fehrmann RS et al. Current smoking-specific gene expression signature in normal bronchial epithelium is enhanced in squamous cell lung cancer. J. Pathol. 218(2), 182–191 (2009).
    • 11. Leitao Filho FS, Takiguchi H, Akata K et al. Effects of inhaled corticosteroid/long-acting beta2-agonist combination on the airway microbiome of patients with chronic obstructive pulmonary disease: a randomized controlled clinical trial (DISARM). Am. J. Respir. Crit. Care Med. 204(10), 1143–1152 (2021).
    • 12. Chua RL, Lukassen S, Trump S et al. COVID-19 severity correlates with airway epithelium–immune cell interactions identified by single-cell analysis. Nat. Biotechnol. 38(8), 970–979 (2020).
    • 13. Deprez M, Zaragosi LE, Truchi M et al. A single-cell atlas of the human healthy airways. Am. J. Respir. Crit. Care Med. 202(12), 1636–1645 (2020).
    • 14. Jaeger B, Schupp JC, Plappert L et al. Airway basal cells show a dedifferentiated KRT17high phenotype and promote fibrosis in idiopathic pulmonary fibrosis. Nat. Commun. 13(1), 5637 (2022).
    • 15. Vieira Braga FA, Kar G, Berg M et al. A cellular census of human lungs identifies novel cell states in health and in asthma. Nat. Med. 25(7), 1153–1163 (2019).
    • 16. Xu K, Shi X, Husted C et al. Smoking modulates different secretory subpopulations expressing SARS-CoV-2 entry genes in the nasal and bronchial airways. Res. Sq. (2021).
    • 17. Zuo WL, Rostami MR, Shenoy SA et al. Cell-specific expression of lung disease risk-related genes in the human small airway epithelium. Respir. Res. 21(1), 200 (2020).
    • 18. Leung JM, Yang CX, Tam A et al. ACE-2 expression in the small airway epithelia of smokers and COPD patients: implications for COVID-19. Eur. Respir. J. 55(5), 2000688 (2020).
    • 19. 10x Genomics. Chromium single cell 3′ reagent kits user guide (v3.1 chemistry) (2020). https://support.10xgenomics.com/permalink/user-guide-chromium-single-cell-3-reagent-kits-user-guide-v31-chemistry
    • 20. Young MD, Behjati S. SoupX removes ambient RNA contamination from droplet-based single-cell RNA sequencing data. GigaScience 9(12), (2020).
    • 21. Li B, Gould J, Yang Y et al. Cumulus provides cloud-based data analysis for large-scale single-cell and single-nucleus RNA-seq. Nat. Methods 17(8), 793–798 (2020).
    • 22. Pegasus Team. Pegasus v1.5.0 (2023). https://github.com/lilab-bcb/pegasus
    • 23. 10x Genomics. Single cell protocols: cell preparation guide (2017). https://cdn.10xgenomics.com/image/upload/v1660261285/support-documents/CG000053_CellPrepGuide_RevC.pdf
    • 24. Reichard A, Asosingh K. Best practices for preparing a single cell suspension from solid tissues for flow cytometry. Cytometry A 95(2), 219–226 (2019).
    • 25. Bajpai R, Lesperance J, Kim M, Terskikh AV. Efficient propagation of single cells Accutase-dissociated human embryonic stem cells. Mol. Reprod. Dev. 75(5), 818–827 (2008).