Skip to main content
  • Research article
  • Open access
  • Published:

Laboratory contamination in airway microbiome studies

Abstract

Background

The low bacterial load in samples acquired from the lungs, have made studies on the airway microbiome vulnerable to contamination from bacterial DNA introduced during sampling and laboratory processing. We have examined the impact of laboratory contamination on samples collected from the lower airways by protected (through a sterile catheter) bronchoscopy and explored various in silico approaches to dealing with the contamination post-sequencing. Our analyses included quantitative PCR and targeted amplicon sequencing of the bacterial 16S rRNA gene.

Results

The mean bacterial load varied by sample type for the 23 study subjects (oral wash>1st fraction of protected bronchoalveolar lavage>protected specimen brush>2nd fraction of protected bronchoalveolar lavage; pā€‰<ā€‰0.001). By comparison to a dilution series of know bacterial composition and load, an estimated 10ā€“50% of the bacterial community profiles for lower airway samples could be traced back to contaminating bacterial DNA introduced from the laboratory. We determined the main source of laboratory contaminants to be the DNA extraction kit (FastDNA Spin Kit). The removal of contaminants identified using tools within the Decontam R package appeared to provide a balance between keeping and removing taxa found in both negative controls and study samples.

Conclusions

The influence of laboratory contamination will vary across airway microbiome studies. By reporting estimates of contaminant levels and taking use of contaminant identification tools (e.g. the Decontam R package) based on statistical models that limit the subjectivity of the researcher, the accuracy of inter-study comparisons can be improved.

Background

The most common method used for studying the bacterial communities of the lower respiratory tract is high throughput amplicon sequencing of the bacterial 16S ribosomal RNA (16S rRNA) marker gene [1]. Some studies use sputum samples [2, 3], with inevitable questions regarding the degree to which the samples are representative of the lower respiratory tract as opposed to contamination from the upper respiratory tract. The emerging gold standard for lower respiratory tract samples is protected bronchoscopy (sampling via a sterile catheter) [4]. However, even with protected bronchoscopy the samples are processed through extensive laboratory workflows that include at minimum steps of bacterial DNA extraction, PCR amplification of the marker gene, and preparation for sequencing. Each step opens up the possibility for the introduction of contaminating bacterial DNA from the laboratory environment, with greatest impact on samples with the lowest bacterial load [5].

Accurate analysis of the lower respiratory tract microbiome will require separate consideration of both of the aforementioned contamination sources - that from the upper respiratory tract introduced during sampling and that introduced during laboratory processing steps. We have previously shown that protected bronchoscopy offers some protection from upper airway contamination [4]. In the current study, we address the issue of contamination from the laboratory.

The impact of laboratory contamination is typically evaluated through the inclusion of negative control samples (NCS) that are processed through all steps of DNA extraction and library preparation for sequencing alongside the study samples. The approach is not perfect as one may expect to find taxa in the NCS that also belong to the bacterial communities of the sampled site. Researchers are thus faced with a difficult decision with regards to what to do with the information acquired from the NCS. Some groups have removed all taxa identified in NCS from their study samples [4, 6, 7]. Others single out taxa they believe likely represent contaminants [8]. Currently bioinformatic tools are being developed that aim to wriggle out the authentic microbiota signal using statistical models [9,10,11], but these have yet to be tested on lower respiratory tract sequencing data (e.g. Decontam [9]).

In the current paper we illustrate an effective workflow for evaluating the quality of lower respiratory tract samples for accurate assessment of bacterial composition. Objectives of the study were i) to determine the influence of contamination on lower respiratory tract samples as a function of bacterial load, ii) to determine the main source of contamination in our laboratory setting and iii) to explore common in silico approaches to dealing with contamination.

Results

In order to establish the bacterial load in protected airway samples collected using different sampling techniques, we included oral washes (OW), two fractions of protected bronchoalveolar lavage (PBAL1 and PBAL2) and protected specimen brushes (PSB) from 23 participants of the MicroCOPD study [12]. The subject characteristics are provided in TableĀ 1.

Table 1 Subject characteristics

Bacterial load varies with sample type

The bacterial load in the four sample types collected per subject was measured by probe based quantitative PCR (qPCR) targeting the bacterial 16S rRNA gene V1ā€‰V2 region. The bacterial load decreased in order OWā€‰>ā€‰PBAL1ā€‰>ā€‰PSBā€‰>ā€‰PBAL2 (pā€‰<ā€‰0.001, non-parametric trend test) (Fig.Ā 1). The mean number of bacteria (Ɨā€‰106/ mL sample) was 34.2 (range 1.4 to 155.8) for OW (nā€‰=ā€‰23); 1.1 (range 1.7ā€‰Ć—ā€‰10āˆ’ā€‰3 to 6.6) for PBAL1 (nā€‰=ā€‰23); 0.7 (range 4.3ā€‰Ć—ā€‰10āˆ’ā€‰3 to 2.8) for PSB (nā€‰=ā€‰20) and 0.5 (range 19.9ā€‰Ć—ā€‰10āˆ’ā€‰3 to 5.1) for PBAL2 (nā€‰=ā€‰23).

Fig. 1
figure 1

Measured bacterial load in procedural samples (OW, PBAL1, PSB and PBAL2). The mean bacterial load in OW samples was approximately 30 fold higher than PBAL1, 50 fold higher than PSB and, 70 fold higher than PBAL2. OW: oral wash (nā€‰=ā€‰23); PBAL1: first fraction of protected BAL from right middle lobe (nā€‰=ā€‰23); PSB: protected specimen brush from right lower lobe (nā€‰=ā€‰20); PBAL2: second fraction of protected BAL from right middle lobe (nā€‰=ā€‰23)

Bacterial load and impact of laboratory contamination

Salter and colleagues [5] have previously illustrated the inverse relationship between the bacterial load in a sample and the influence of contamination on the bacterial community readout. Once we had established that the bacterial load varied with sampling technique (Fig. 1), we questioned whether the differences in bacterial load for each of the patient samples would also reflect differences in susceptibility to laboratory contamination. Using the Salter approach [5], we estimated the degree of contamination as a function of bacterial load (Fig.Ā 2), and translated this to an estimate of contamination in the procedural samples (OW, PBAL, PSB). Using quantitative PCR we determined that the initial Salmonella sample had a concentration of 107 bacterial cells/mL. As expected the oral wash samples having a high bacterial load (mean of approximately 107 bacterial cells/mL), will not be greatly impacted by contamination. Samples from the lungs (PBAL, PSB) fell between dilution 2 and 3 (Fig. 2), with contamination representing 10ā€“50% of the bacterial community readout. The impact of varying number of PCR cycles was low (Fig. 2).

Fig. 2
figure 2

Estimate of contaminant levels in ten-fold dilution series of Salmonella (SDS). The major operational taxonomic units (OTUs) observed in the initial Salmonella sample (10^7 bacteria/mL) were assigned to f__Enterobacteriaceae;g__. Using the NCBI nucleotide BLAST tool we confirmed that these OTUs (OTU821080, OTU813457 and OTU813217) matched to the genus Salmonella. With each successive dilution, the relative abundance of f__Enterobacteriaceae;g__ decreased. By dilution 3 (45 PCR cycles), the percentage had reduced to 47.83%. For comparison, PCR amplification of the 16S rRNA gene was performed at both 30 and 45ā€‰cycles for all SDS samples. The control is a sample of PCR water processed through steps of PCR and sequencing alongside the SDS samples. Taxonomic rank is described using prefixes (f__: family, g__: genus)

Monitoring procedural contamination

Having learned that contaminating bacterial DNA likely represents a substantial proportion (10ā€“50%) of the sequencing output for the lower airway samples in our study, we attempted to identify the main contamination source. We performed ten simulated bronchoscopy procedures (no patient) over two days to capture the environmental contaminants that may have been introduced during sampling.

All procedural control samples were sequenced together on the same sequencing run (Run A). Additional control samples were sequenced on a second run (Run B) and included samples of molecular grade water that were processed through the DNA extraction protocol without the introduction of PBS. Although sequenced on a separate sequencing run (Run B), the molecular grade water samples would indicate whether the PBS was the main source of contamination. A sample of molecular grade water that was not processed through the DNA extraction protocol (PCR water) was also included on both sequencing runs (Run A and B). This later sample would reflect contamination introduced during PCR and sequencing steps without interference from contamination introduced during sampling and DNA extraction steps.

The total number of sequences obtained from the procedural control samples (Run A) after quality filtering and chimera removal was 4.8ā€‰Ć—ā€‰106. The mean number of sequences and operational taxonomic units (OTUs) obtained from the procedural controls were for phosphate buffered saline (nā€‰=ā€‰10): 64,745 sequences (123 OTUs); catheter rinse (nā€‰=ā€‰10): 98,379 sequences (131 OTUs); protected specimen brushes (nā€‰=ā€‰10): 106,853 sequences (132 OTUs); bronchoscope rinse (nā€‰=ā€‰10): 109,765 sequences (134 OTUs); cryotube (nā€‰=ā€‰9): 115,633 sequences (138 OTUs). The number of sequences obtained from the PCR water control sequenced on the same run (Run A) was lower than for the procedural control samples with only 43,433 sequences and 65 OTUs, suggesting that contamination was predominantly introduced prior to PCR steps of library preparation. The procedural control samples (Run A) showed a similar taxonomic distribution that was quite distinct from that of the PCR water sample (Run A) (Fig.Ā 3). This indicated that contamination was either introduced with the phosphate buffered saline used for collection of all samples or during DNA extraction steps.

Fig. 3
figure 3

Distribution of operational taxonomic units (OTUs) in procedural controls and PCR water samples. An OTU belonging to the genera Ralstonia dominated the procedural control samples with an average relative abundance of 51.81% in scope rinse (nā€‰=ā€‰10), 54.33% in catheter rinse (nā€‰=ā€‰10), 55.36% in cryotube (nā€‰=ā€‰9), 52.82% in protected specimen brushes (nā€‰=ā€‰10) and 54.93% in phosphate buffered saline (nā€‰=ā€‰10). The same Ralstonia OTU also dominated the molecular grade water samples (nā€‰=ā€‰3) at an average relative abundance of 29.42%. The PCR water control sample was dominated by Rhizobium (38.11%), Anaerobacillus New Reference OTU 110 (20.69%) and Delftia (10.65%) in run A and Anaerobacillus New Reference OTU 110 (32.93%), Anaerobacillus OTU 622288 (24.04%) and Delftia (10.68%) in run B. Taxonomic rank is described using prefixes (o__: order, f__: family, g__: genus). Data unrarefied

To differentiate between PBS and DNA extraction as contamination sources, we compared the molecular grade water samples (Run B) to the corresponding PCR water sample sequenced on the same run. The molecular grade water (nā€‰=ā€‰3) (Run B) contained a mean number of 124,941 sequences and 107 OTUs, whereas the PCR water (Run B) contained 126,103 sequences and only 39 OTUs. Importantly, the taxonomic profile of the molecular grade water (Run B) resembled that of the procedural control samples (Run A), whereas the PCR water did not, indicating that the main source of contamination was the DNA extraction kit (Fig. 3).

Exploring in silico approaches to dealing with contamination in LRT samples

We began our analyses by looking at how the top 20 OTUs present in NCS were distributed in the procedural samples (OW, PBAL, PSB) in our 23 subjects (Fig.Ā 4). The NCS were dominated by an OTU that mapped to the family Enterobacteriaceae. The Ralstonia OTU that dominated the procedural controls (Fig. 3) was the fourth most abundant OTU in the NCS with an average relative abundance of just 5.45%. This likely reflects differences in contamination introduced from different lots of the FastDNA Spin Kit [5]. An OTU assigned to the Streptococcus genus was found in NCS at a relative abundance of just 1.51%; the same OTU was a major OTU in patient OW, PBAL and PSB samples. This is most likely not a contaminant and may be an important component of the bacterial lung microbiota. For a detailed presentation of the Streptococcus OTUs found in PSB and NCS samples, see AdditionalĀ fileĀ 1: Figure S1 and AdditionalĀ fileĀ 2: Figure S2.

Fig. 4
figure 4

Distribution of the 20 most abundant operational taxonomic units (OTUs) observed in negative control samples (NCS). The NCS were dominated by OTU 759061 assigned to the family Enterobacteriaceae (20.93%), OTU 4389128 assigned to a genus within the class ML635J-21 (16.31%), OTU 437105 and New. Reference OTU 133 both assigned to the genus Ralstonia (8.30 and 5.45%, respectively). Taxonomic rank is described using prefixes (c__: class, o__: order, f__: family, g__: genus). Data presented as the average relative abundance. Data unrarefied

Common in silico approaches to dealing with contamination include i) leaving the samples intact (i.e. do nothing), ii) removing all OTUs seen in NCS, and iii) correction based on statistical models (i.e. the Decontam R package). We next examined how the application of each approach would impact the taxonomic profiles of the procedural samples in our study (Fig.Ā 5).

Fig. 5
figure 5

Taxonomic distribution in procedural samples when different approaches to dealing with contamination have been applied. When negative control sample (NCS) operational taxonomic units (OTUs) are kept, the Streptococcus genus dominated the procedural samples with an average relative abundance of 31.66% in oral wash (OW) (nā€‰=ā€‰23), 27.95% in protected bronchoalveolar lavage (PBAL) (nā€‰=ā€‰23) and 22.27% in protected specimen brushes (PSB) (nā€‰=ā€‰23). With the removal of NCS OTUs, the Streptococcus genus no longer dominated the procedural samples and was present at an average relative abundance of 4.80% in OW (nā€‰=ā€‰23), 6.12% in PBAL (nā€‰=ā€‰23) and 7.60% in PSB (nā€‰=ā€‰23). With the removal of OTUs identified as contaminants in Decontam (methodā€‰=ā€‰ā€œeitherā€, thresholdā€‰=ā€‰0.5), the Streptococcus genus again dominated the samples, with an average relative abundance of 32.52% in OW (nā€‰=ā€‰23), 34.40% in PBAL (nā€‰=ā€‰23) and 35.08% in PSB (nā€‰=ā€‰23). Taxonomic rank is described using prefixes (c__: class, o__: order, f__: family, g__: genus). Data unrarefied

When leaving the procedural samples intact, the Streptococcus genus dominated all sample types. With the removal of OTUs seen in NCS, the relative abundance of the Streptococcus genus was significantly reduced in all sample types (Fig. 5), as was predicted from Fig. 4. With removal of OTUs identified as contaminants using Decontam [9], the Streptococcus genus again dominated the procedural samples. This approach thus appeared to provide a good balance between removing all OTUs found in the NCS and leaving intact OTUs present in both NCS and procedural samples. Comparison of the frequency-based distribution plots for the top 4 OTUs observed in NCS and the Streptococcus OTU (Fig.Ā 6), visually illustrate how Decontam (here frequency-based method) is able to differentiate between a contaminant OTU and a non-contaminant OTU.

Fig. 6
figure 6

Decontam frequency distribution plots distinguish contaminants from non-contaminants. A frequency distribution plot generated from samples with varying DNA concentration indicates whether a particular sequence fits the Decontam contaminant (red line) or non-contaminant (black stippled line) model. The first four plots represent the top four operational taxonomic units (OTUs) observed in negative control samples (NCS): OTU 759061 is assigned to the family Enterobacteriaceae; OTU 4389128 is assigned to a genus within the class ML635J-21; OTU 437105 and OTU New. Reference OTU 133 are both assigned to the genus Ralstonia. The final plot represents the Streptococcus OTU 1082539 that most likely is not a contaminant, although present among the top 20 OTUs found in NCS. Its frequency distribution pattern more closely fits the Decontam non-contaminant model in contrast to the others

Decontam performance test on the Salmonella dilution series (SDS)

In the Decontam introduction paper [9], the authors illustrate how Decontam is able to diminish the contaminant signal from the serially diluted Salmonella datasets published in the Salter paper [5]. As our study also included a Salmonella dilution series (SDS), we were able to test the Decontam package tools on sequencing data generated in the context of our laboratory setting after processing through our chosen bioinformatic pipeline.

The SDS in our study included seven samples of a successively ten-fold diluted Salmonella monoculture and a PBS negative control sample that went through DNA extraction and sequencing steps alongside the SDS (Fig.Ā 7). As library preparation for sequencing of the SDS was performed at both 30 and 45 PCR cycles and the impact of varying number of PCR cycles was low (Fig. 2), the sequencing output for both sample sets were used as input in the Decontam analyses. We also included a PCR water control sample that was sequenced on the same sequencing run.

Fig. 7
figure 7

Taxonomic distribution in Salmonella dilution series (SDS). The taxonomic profile of the SDS samples (amplified using 45 PCR cycles) before removal of OTUs identified as contaminants in Decontam. Taxonomic rank is described using prefixes (f__: family, g__: genus). Data unrarefied

Using the isContaminant function in the Decontam R package, we compared three methods for identification of contaminant OTUs including i) the prevalence-based method, ii) the frequency-based method and iii) the either method. In the prevalence-based method, an OTU is marked as a contaminant based on a comparison of how often the OTU is observed in negative control samples compared to the samples under study. For testing the approach on the SDS, the final two samples in the SDS were assigned as negative control samples together with PBS and PCR water samples (as conducted by Decontam developers when testing the approach on the Salter datasetĀ [13]). FigureĀ 8 shows the taxonomic profile of the SDS samples after removal of contaminant OTUs identified using the prevalence-based approach. The impression was that many small OTUs were removed. In the frequency-based approach, the labelling of an OTU as a contaminant is based on the correlation between the DNA concentration measurements made for samples during steps of library preparation (in our lab using the Qubit instrument) and the relative abundance of the OTU across samples. FigureĀ 9 shows the taxonomic profile of the SDS samples after removal of contaminant OTUs identified using the frequency-based approach. The impression was that the frequency-based approach removed fewer but more abundant OTUs compared to the prevalence-based approach. In the final approach tested in Decontam (ā€œeitherā€), all OTUs marked as contaminants by either the prevalence or frequency-based methods are removed (Fig.Ā 10).

Fig. 8
figure 8

Taxonomic distribution in Salmonella dilution series (SDS) after removal of Decontam contaminants (prevalence-based). 109 out of 235 operational taxonomic units (OTUs) in the SDS dataset were identified as contaminants and removed (user defined thresholdā€‰=ā€‰0.5). At the default threshold, only 34 out of 235 OTUs in the dataset were identified as contaminants (figure not drawn). Taxonomic rank is described using prefixes (f__: family, g__: genus). Data unrarefied

Fig. 9
figure 9

Taxonomic distribution in Salmonella dilution series (SDS) after removal of Decontam contaminants (frequency-based). 58 out of 235 operational taxonomic units (OTUs) in the dataset were identified as contaminants and removed (user defined thresholdā€‰=ā€‰0.5). At the default threshold, only 9 out of 235 OTUs in the dataset were identified as contaminants (figure not drawn). Taxonomic rank is described using prefixes (c__: class, o__: order, f__: family, g__: genus). Data unrarefied

Fig. 10
figure 10

Taxonomic distribution in Salmonella dilution series (SDS) after removal of Decontam contaminants (approach either). 136 out of 235 OTUs in the dataset were identified as contaminants and removed (user defined thresholdā€‰=ā€‰0.5). Taxonomic rank is described using prefixes (f__: family, g__: genus). Data unrarefied

Of the three approaches tested in Decontam, the ā€œeitherā€ method was able to most effectively remove the contaminant signal from the bacterial community profiles of the samples; even in the most diluted sample over 50% of the sequences mapped to the Salmonella genus. Of concern is however that the PBS sample also consisted of over 50% Salmonella. Also present in the PBS sample was oral/lung specific genera including Veillonella, Streptococcus and Neisseria that are obvious contaminants from the procedural samples sequenced on the same run. The number of reads in the PBS sample after processing in Decontam was only 32. Therefore we learn that although effective, removal of contaminant OTUs identified in Decontam may also lead to the magnification of another type of noise in the sequencing data ā€“ particularly that from cross sample contamination during library preparation or index misassignment during MiSeq sequencing.

Discussion

In the current paper we illustrate an effective workflow for evaluating the quality of lower airway samples for amplicon-based analysis of bacterial composition. Our results show that the low bacterial load in samples from the lungs make them vulnerable to bacterial DNA contamination, which in our study mainly originated from DNA extraction kits. Even with contaminants representing an estimated 10ā€“50% of the sequencing output for these samples, we demonstrate that most of the contaminating signal can be removed post sequencing using recently developed bioinformatic approaches.

Through the processing and sequencing of a serially diluted culture of Salmonella [5], we were able to define the threshold bacterial load for which contamination would begin to dominate the bacterial profile in our samples. At an input of between 10^3 and 10^4 Salmonella/mL, we observed that contaminants constituted more than 50% of the bacterial profile of the sample. The use of alternative protocols for sample processing and sequencing can slide this defined threshold of bacterial load up or down and should therefore be determined independently in separate studies. Biesbroek et al. [14] for example show in their study how the choice of DNA extraction kit will affect the DNA yield and in turn the placement of samples above or below a defined threshold of bacterial load for which contamination becomes a problem. Despite differences in laboratory protocols, our results are in agreement with Salter and colleagues [5] who in their study also recommend an input of more than 10^3ā€“10^4 bacterial cells. The concordance of our results may partially be explained by the use of a DNA extraction kit from the same manufacturer (FastDNA Spin Kit, MP Biomedicals).

Using the Salmonella dilution series as a reference we were able to determine the degree of laboratory contamination in the various sample types (OW, PBAL1, PBAL2, PSB) collected from participants in the MicroCOPD study. The average bacterial load in the samples acquired from the lungs was highest for PBAL1 samples (10^6 bacteria/mL) and approximately an order of magnitude lower for PSB and PBAL2 samples. This could mean that the first lavage fraction harvests a larger portion of the resident microbiota, but also a dilution effect, as lavage yield tends to increase in the second fraction. We used a sterile inner catheter for lavage sampling, to minimize contamination from BAL, something no other study has done to our knowledge. It is however possible that the first fraction of lavage (PBAL1) is more susceptible to contamination from the upper airways during sampling compared to PBAL2 and PSB samples [4]. Thus, the question remains as to whether PBAL1 with its higher bacterial load is a more representative sample compared to PBAL2 and PSB samples or if we are simply swapping contamination sources (contaminating bacterial DNA introduced from the upper airways during sampling versus contaminating bacterial DNA introduced during laboratory processing steps). The optimal sample type may thus be a question of which contamination source is easiest to identify and remove post sequencing.

Through the sequencing of procedural control samples and PCR negative control samples that were not processed through the DNA extraction protocol, we were able to trace the main source of contamination back to the DNA extraction kit. Our findings are in agreement with several other studies [5, 15, 16]. The difference in the microbiota readout for the procedural control samples and the negative control samples are likely explained by differences in lot number for the DNA extraction kits. Salter and colleagues report differences in contaminant profiles for three replicates of SDS extracted using different lots of the FastDNA Spin Kit for soil; similar to our results they also found that one SDS replicate was dominated by unclassified Enterobacteriaceae.

Publications such as that by Salter and colleagues have led to an increased awareness of the effects of contamination on microbiome studies of low biomass samples [5, 16]. Most studies now process negative control samples that allow for monitoring of the contaminant signal introduced from the laboratory. However, the inclusion of NCS only partly addresses the issue. In our study for example, we recognized that a major Streptococcus OTU found in procedural samples (OW, PBAL, PSB) was also among the top 20 most abundant OTUs found in NCS. A comparison of the relative abundance of the Streptococcus OTU in procedural samples and NCS indicated that the OTU was likely not a contaminant. However, the question of where to draw the line with regards to a set abundance threshold for which an OTU should be identified as a contaminant or not is not always as straightforward. The Decontam package in R has been developed to identify contaminants using statistical models [9]. The Decontam developers demonstrate the accuracy of their approach on the Salmonella dilution series datasets generated in the Salter publication. We show in the context of our laboratory setting that Decontam is efficient at removing the contaminant signal from the SDS also in our study. Using Decontam we were also able to confirm the identity of the Streptococcus OTU found in both procedural samples and the NCS as a non-contaminant.

We acknowledge that our study does not address all issues related to bacterial load in microbiome sequencing data. The serial diluted Salmonella monoculture does not provide insight into the effects of bacterial load on the relative abundance of bacteria in a more complex microbiota sample. Biesbroek et al. [14] show in their study examining the microbiota of a serially diluted saliva sample, an increase in the relative abundance of Proteobacteria and Firmicutes and a decrease in Bacteroidetes across the dilution series. Proteobacteria likely reflect contaminants as has been suggested in several papers [14, 17], again illustrating the inverse relationship between bacterial load and the influence of contamination as observed in our study. The observed increase in relative abundance of Firmicutes and concurrent decrease in Bacteroidetes is however of concern, as these phyla hold members often detected in studies of the lung microbiome (e.g. Veillonella and Prevotella). The field would benefit from studies addressing the potential effects of bacterial load on the measured relative abundance of taxa in a more complex sample, particularly those that are suspect core lung microbiota members. Secondly, we did not quantify the amount of human DNA in the procedural samples. The presence of human DNA may affect the efficiency of the qPCR reaction [16], and thereby also the accuracy of the direct comparison to the SDS. Studies evaluating the impact of contamination might consider quantification of human DNA for an even more accurate estimate of contamination.

Conclusions

Measured amounts of bacteria will vary in lower airway samples collected with different bronchoscopic sampling techniques (e.g. PBAL1, PBAL2, PSB in the current study). These differences combined with the inverse relationship between bacterial load and bacterial DNA contamination will render some sampling modalities dominated by contaminating taxa.

Differences in protocols for sampling, laboratory processing and bioinformatics analysis across studies will require investigators to evaluate the impact of contamination in the context of their own laboratory setting. We encourage investigators to report an estimate of the degree of contamination in their datasets defined against a sample of known bacterial load as exemplified in the current study. We further suggest the use of contaminant identification tools (e.g. Decontam) based on statistical models for the objective removal of laboratory contaminants in lung microbiome sequencing data. Such measures will enable more accurate inter-study comparisons and may also resolve discrepancies between studies that have likely impeded understanding the potential relationship between microbiota and its role in chronic lung diseases.

Methods

Study samples

Study subjects (nā€‰=ā€‰23) were chosen from the Bergen COPD Microbiome Study (short name ā€œMicroCOPDā€) [12], to give an equal representation of healthy (nā€‰=ā€‰9) and diseased (asthma (nā€‰=ā€‰4), COPD (nā€‰=ā€‰10)) states. Details on data collection and the bronchoscopy procedures have been previously published [4, 12]. Briefly, adult subjects recruited from Western Norway with and without obstructive lung disease, underwent voluntary bronchoscopies between 2013 and 2015. All subjects were examined in the stable state, not having received antibiotics at least 2ā€‰weeks prior to the procedure. All bronchoscopies were performed by experienced chest physicians at the outpatient clinic at the Department of Thoracic Medicine, Haukeland University Hospital. The regional ethical committee (REK-Nord, case # 2011/1307) approved the study, and all patients gave written informed consent.

Sample types acquired per patient included the first and second fraction of 2ā€‰Ć—ā€‰50ā€‰mL bronchoalveolar lavage (PBAL1 and PBAL2) sampled through a sterile inner catheter (Plastimed Combicath, Le Plessis Bouchard, France) of the bronchoscope while the scope itself was wedged in the right middle lobe, and three protected specimen brushes subsequently sampled from the right lower lobe (rPSB), an oral wash (OW), and a negative control sample (NCS). Additional procedural control samples were collected after ten simulated bronchoscopy procedures (no patient) carried out over two days; samples included a bronchoscope rinse (BR), a catheter rinse (CR), a protected specimen brush (PSB), a sample of phosphate buffered saline (PBS) transferred to a cryotube (CT) and a sample of PBS used for collection of all samples. The PBS used for sample collection was sterilized by sterile filtration (0.22ā€‰Ī¼m) and autoclaving at 121ā€‰Ā°C for 15ā€‰min. To study the relationship between bacterial load and the influence of contaminating bacterial DNA in our laboratory setting [5], we included a ten-fold dilution series of Salmonella enterica serovar Typhimurium (ATCC 14028) (ATCC, Manassas, VA, USA) (SDS).

Bacterial DNA extraction using enzymatic and mechanical lysis steps

Samples were treated with lytic enzymes mutanolysin, lysozyme and lysostaphin (all from Sigma-Aldrich, St. Louis, MO, USA) and subsequently processed through the FastDNA Spin Kit (MP Biomedicals, LLC, Solon, OH, USA) following the manufacturerā€™s instructions. Procedural samples were processed using different lots of the DNA extraction kit (#79113, #84562, #57212, #62903). The procedural controls and the SDS were processed using a kit of same lot number (#93678). The sample volume used as input varied with sample type (for procedural samples: 450ā€‰Ī¼l for PSB and NCS and 1800ā€‰Ī¼l for OW, PBAL1, PBAL2; for procedural control samples: 450ā€‰Ī¼l for PBS and CT, 550ā€‰Ī¼l for PSB and 1800ā€‰Ī¼l for BR and CR; for samples in the SDS: 500ā€‰Ī¼l). DNA was eluted in a total volume of 100ā€‰Ī¼l.

Quantification of bacterial load by quantitative PCR (qPCR)

The bacterial load in the samples was determined by probe-based qPCR targeting the bacterial 16S rRNA gene (region V1ā€‰V2) using forward primer 5ā€²-AGAGTTTGATCCTGGCTCAG-3ā€², reverse primer 5ā€²-CTGCTGCCTYCCGTA-3ā€² and probe 5ā€²-6-FAM-TAACACATGCAAGTCGA-BHQ-1-3ā€² (locked nucleic acid bases are underlined; 6-FAM: 6-carboxyfluorescein; BHQ-1: Black Hole Quencher-1) [7, 18,19,20]. PCR reactions were carried out using the following cycling conditions: an initial cycle at 95ā€‰Ā°C for 5ā€‰min followed by 45ā€‰cycles of 95ā€‰Ā°C for 5ā€‰s, 60ā€‰Ā°C for 20ā€‰s and 72ā€‰Ā°C for 10ā€‰s and a final extension cycle of 72ā€‰Ā°C for 2ā€‰min. A standard curve was constructed from genomic DNA from E. coli strain JM109 (Zymo Research, Irvine, CA, USA).

MiSeq sequencing of the bacterial 16S rRNA gene

The bacterial composition in the samples was determined by paired-end sequencing of the 16S rRNA gene (region V3ā€‰V4) following instructions provided in the Illumina 16S Metagenomic Sequencing Library Preparation guide (Part no. 15044223 Rev. B). PCR cycling conditions were modified from the commercial protocol and consisted of an initial cycle at 95ā€‰Ā°C for 3ā€‰min followed by 45ā€‰cycles of 95ā€‰Ā°C for 30ā€‰s, 55ā€‰Ā°C for 30ā€‰s, 72ā€‰Ā°C for 30ā€‰s and a final extension cycle at 72ā€‰Ā°C for 5ā€‰min.

Bioinformatic sequence processing steps

Bioinformatic sequence processing steps were performed using tools provided within the Quantitative Insights into Microbial Ecology (QIIME) bioinformatic package, version 1.9.1. In short, raw sequences were retrieved from the MiSeq sequencer in the form of demultiplexed forward and reverse fastq files (paired end reads). Primer sequences were trimmed off and forward and reverse reads joined. Chimera sequences identified using the VSEARCH program [21] were subsequently removed. Remaining sequences were grouped into open-reference operational taxonomic units (OTUs) using UCLUST [22] and the GreenGenes reference database (v.13.8) [23]. Small OTUs, defined as those containing less than 0.005% of the total sequence count in the dataset were then filtered out [24]. Taxonomy was assigned to OTUs using the naĆÆve bayesian RDP Classifier [25] together with the GreenGenes reference database (v.13.8) [23]. The resulting OTU table displaying the sequence count in each OTU for each sample was the starting point for all subsequent analyses. The QIIME commands used for generating the working OTU table are provided in the AdditionalĀ fileĀ 3: Supplementary Methods.

In silico contaminant identification and removal

Two approaches to contaminant identification and subsequent removal were tested. In the first approach contaminant OTUs were identified through their presence in NCS. NCS OTUs were filtered out from the procedural samples (OW, PSB, PBAL) collected under the same procedure using QIIME commands (illustrated in the supplementary methods). In the second approach, contaminant OTUs were identified based on statistical models using the Decontam package [9] in R. Contaminant OTUs identified using the Decontam isContaminant function (methodā€‰=ā€‰either, user defined thresholdā€‰=ā€‰0.5) were filtered out of the main OTU working table using QIIME commands.

For greater details on study design, sample collection, preparation of Salmonella samples, DNA extraction, qPCR, 16S rRNA gene sequencing and bioinformatics, please see the Additional file 3: Supplementary Methods.

Availability of data and materials

The fastq files and metadata needed to rerun the analyses performed in the current study will be available in the DRYAD repository upon publication (doi:https://0-doi-org.brum.beds.ac.uk/10.5061/dryad.1v92t8b). Bioinformatics analyses steps are described in the Additional file 3: Supplementary Methods. A detailed description of the protocols and laboratory materials used in the MicroCOPD Study are available at dx.doi.org/10.17504/protocols.io.2sygefw.

Abbreviations

COPD:

Chronic obstructive pulmonary disease

NCS:

Negative control sample

OTU:

Operational taxonomic unit

OW:

Oral wash

PBAL:

Protected bronchoalveolar lavage

PSB:

Protected specimen brush

QIIME:

Quantitative Insights into Microbial Ecology

qPCR:

Quantitative PCR

References

  1. Hilty M, Burke C, Pedro H, Cardenas P, Bush A, Bossley C, et al. Disordered microbial communities in asthmatic airways. PLoS One. 2010;5:e8578.

    ArticleĀ  Google ScholarĀ 

  2. Tangedal S, Aanerud M, GrĆønseth R, Drengenes C, Wiker HG, Bakke PS, et al. Comparing microbiota profiles in induced and spontaneous sputum samples in COPD patients. Respir Res. 2017;18:164.

    ArticleĀ  Google ScholarĀ 

  3. Leitao Filho FS, Alotaibi NM, Ngan D, Tam S, Yang J, Hollander Z, et al. Sputum microbiome is associated with 1-year mortality following COPD hospitalizations. Am J Respir Crit Care Med. 2018. https://0-doi-org.brum.beds.ac.uk/10.1164/rccm.201806-1135OC.

    ArticleĀ  Google ScholarĀ 

  4. GrĆønseth R, Drengenes C, Wiker HG, Tangedal S, Xue Y, HusebĆø GR, et al. Protected sampling is preferable in bronchoscopic studies of the airway microbiome. ERJ Open Res. 2017;3.

    ArticleĀ  Google ScholarĀ 

  5. Salter SJ, Cox MJ, Turek EM, Calus ST, Cookson WO, Moffatt MF, et al. Reagent and laboratory contamination can critically impact sequence-based microbiome analyses. BMC Biol. 2014;12:87.

    ArticleĀ  Google ScholarĀ 

  6. Einarsson GG, Comer DM, McIlreavey L, Parkhill J, Ennis M, Tunney MM, et al. Community dynamics and the lower airway microbiota in stable chronic obstructive pulmonary disease, smokers and healthy non-smokers. Thorax. 2016;71:795ā€“803.

    ArticleĀ  CASĀ  Google ScholarĀ 

  7. Dickson RP, Erb-Downward JR, Freeman CM, Walker N, Scales BS, Beck JM, et al. Changes in the lung microbiome following lung transplantation include the emergence of two distinct Pseudomonas species with distinct clinical associations. PLoS One. 2014;9. https://0-doi-org.brum.beds.ac.uk/10.1371/journal.pone.0097214.

    ArticleĀ  Google ScholarĀ 

  8. Pragman AA, Lyu T, Baller JA, Gould TJ, Kelly RF, Reilly CS, et al. The lung tissue microbiota of mild and moderate chronic obstructive pulmonary disease. Microbiome. 2018;6:7.

    ArticleĀ  Google ScholarĀ 

  9. Davis NM, Proctor DM, Holmes SP, Relman DA, Callahan BJ. Simple statistical identification and removal of contaminant sequences in marker-gene and metagenomics data. Microbiome. 2018;6:226.

    ArticleĀ  Google ScholarĀ 

  10. Jervis-Bardy J, Leong LEX, Marri S, Smith RJ, Choo JM, Smith-Vaughan HC, et al. Deriving accurate microbiota profiles from human samples with low bacterial content through post-sequencing processing of Illumina MiSeq data. Microbiome. 2015;3:19.

    ArticleĀ  Google ScholarĀ 

  11. Lazarevic V, GaĆÆa N, Girard M, Schrenzel J. Decontamination of 16S rRNA gene amplicon sequence datasets based on bacterial load assessment by qPCR. BMC Microbiol. 2016;16:73.

    ArticleĀ  Google ScholarĀ 

  12. GrĆønseth R, Haaland I, Wiker HG, Martinsen EMH, Leiten EO, HusebĆø G, et al. The Bergen COPD microbiome study (MicroCOPD): rationale, design, and initial experiences. Eur Clin Respir J. 2014;1:26196.

    ArticleĀ  Google ScholarĀ 

  13. Davis NM, Proctor D, Holmes SP, Relman DA, Callahan BJ. Simple statistical identification and removal of contaminant sequences in marker-gene and metagenomics data. bioRxiv. 2018. https://0-doi-org.brum.beds.ac.uk/10.1101/221499.

  14. Biesbroek G, Sanders EAM, Roeselers G, Wang X, Caspers MPM, Trzciński K, et al. Deep sequencing analyses of low density microbial communities: working at the boundary of accurate microbiota detection. PLoS One. 2012;7:e32942.

    ArticleĀ  CASĀ  Google ScholarĀ 

  15. Lauder AP, Roche AM, Sherrill-Mix S, Bailey A, Laughlin AL, Bittinger K, et al. Comparison of placenta samples with contamination controls does not provide evidence for a distinct placenta microbiota. Microbiome. 2016;4:29.

    ArticleĀ  Google ScholarĀ 

  16. Glassing A, Dowd SE, Galandiuk S, Davis B, Chiodini RJ. Inherent bacterial DNA contamination of extraction and sequencing reagents may affect interpretation of microbiota in low bacterial biomass samples. Gut Pathog. 2016;8. https://0-doi-org.brum.beds.ac.uk/10.1186/s13099-016-0103-7.

  17. Willner D, Daly J, Whiley D, Grimwood K, Wainwright CE, Hugenholtz P. Comparison of DNA extraction methods for microbial community profiling with an application to pediatric Bronchoalveolar lavage samples. PLoS One. 2012;7:e34605.

    ArticleĀ  CASĀ  Google ScholarĀ 

  18. Charlson ES, Bittinger K, Haas AR, Fitzgerald AS, Frank I, Yadav A, et al. Topographical continuity of bacterial populations in the healthy human respiratory tract. Am J Respir Crit Care Med. 2011;184:957ā€“63.

    ArticleĀ  Google ScholarĀ 

  19. Dickson RP, Erb-Downward JR, Freeman CM, McCloskey L, Falkowski NR, Huffnagle GB, et al. Bacterial topography of the healthy human lower respiratory tract. mBio. 2017;8:e02287-16.

    ArticleĀ  Google ScholarĀ 

  20. Bassis CM, Erb-Downward JR, Dickson RP, Freeman CM, Schmidt TM, Young VB, et al. Analysis of the upper respiratory tract microbiotas as the source of the lung and gastric microbiotas in healthy individuals. mBio. 2015;6:e00037-15.

    ArticleĀ  Google ScholarĀ 

  21. Rognes T, Flouri T, Nichols B, Quince C, MahƩ F. VSEARCH: a versatile open source tool for metagenomics. PeerJ. 2016;4. https://0-doi-org.brum.beds.ac.uk/10.7717/peerj.2584.

    ArticleĀ  Google ScholarĀ 

  22. Edgar RC. Search and clustering orders of magnitude faster than BLAST. Bioinforma Oxf Engl. 2010;26:2460ā€“1.

    ArticleĀ  CASĀ  Google ScholarĀ 

  23. McDonald D, Price MN, Goodrich J, Nawrocki EP, DeSantis TZ, Probst A, et al. An improved Greengenes taxonomy with explicit ranks for ecological and evolutionary analyses of bacteria and archaea. ISME J. 2012;6:610ā€“8.

    ArticleĀ  CASĀ  Google ScholarĀ 

  24. Bokulich NA, Subramanian S, Faith JJ, Gevers D, Gordon JI, Knight R, et al. Quality-filtering vastly improves diversity estimates from Illumina amplicon sequencing. Nat Methods. 2013;10:57ā€“9.

    ArticleĀ  CASĀ  Google ScholarĀ 

  25. Wang Q, Garrity GM, Tiedje JM, Cole JR. NaĆÆve Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Env Microbiol. 2007;73:5261ā€“7.

    ArticleĀ  CASĀ  Google ScholarĀ 

Download references

Acknowledgements

The authors wish to thank Marit Aardal, Kristina Apalseth, Hildegunn Bakke Fleten, Ane Aamli Gagnat, Ingvild Haaland, Tuyen Thi Van Hoang, Gunnar HusebĆø, Kristel Knudsen, Sverre Lehmann, Lise ƘstgĆ„rd Monsen, Randi Sandvik, Ƙistein Svanes for their contributions in the data collection and/or analyses.

Funding

The MicroCOPD study was funded by unrestricted grants and fellowships from Helse Vest, Bergen Medical Research Foundation, the Endowment of timber merchant A. Delphin and wife through the Norwegian Medical Association and GlaxoSmithKline through the Norwegian Respiratory Society. The funding bodies had no role in the design of the study, data collection and analysis, interpretation of data, or in writing the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

TME, RN, HGW, EN and TK participated in the planning and collection of procedural samples in the MicroCOPD study. HGW planned the sequencing analyses. CD, TK, EN, TME and RN participated in planning and collection of procedural control samples. CD and TK performed DNA extraction and library preparation for sequencing. CD performed qPCR, bioinformatics analyses and drafted the manuscript. RN and TME participated in bioinformatics analyses and drafting of the manuscript. All authors participated in the revision of the manuscript and approved the final version for publication.

Corresponding author

Correspondence to Christine Drengenes.

Ethics declarations

Ethics approval and consent to participate

The study was approved by the Regional Committees for Medical and Health Research Ethics (REK-Nord, case # 2011/1307) and was conducted in accordance with the Declaration of Helsinki. All study subjects signed informed consent forms.

Consent for publication

Not applicable.

Competing interests

CD, HGW, TK, EN: The authors declare that they have no competing interests.

TME: Reports bursary from Boehringer Ingelheim for educational meetings within the last three years, unrelated to the current study.

RN: Reports grants from GlaxoSmithKline, during the conduct of the study; grants from Boehringer Ingelheim, grants and personal fees from AstraZeneca, grants from Novartis, grants from Boehringer Ingelheim, personal fees from GlaxoSmithKline, outside the submitted work.

Additional information

Publisherā€™s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Additional files

Additional file 1:

Figure S1. Distribution of Streptococcus OTUs in Protected Specimen Brush (PSB) samples (n=23). (PDF 8 kb)

Additional file 2:

Figure S2. Distribution of Streptococcus OTUs in Negative Control Samples (NCS) (n=23). (PDF 8 kb)

Additional file 3:

Supplementary Methods. This file provides a detailed description of protocols for sample collection, preparation of Salmonella samples, DNA extraction, qPCR, 16S rRNA gene sequencing and bioinformatics. (DOCX 240 kb)

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Drengenes, C., Wiker, H.G., Kalananthan, T. et al. Laboratory contamination in airway microbiome studies. BMC Microbiol 19, 187 (2019). https://0-doi-org.brum.beds.ac.uk/10.1186/s12866-019-1560-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://0-doi-org.brum.beds.ac.uk/10.1186/s12866-019-1560-1

Keywords