Register      Login
Microbiology Australia Microbiology Australia Society
Microbiology Australia, bringing Microbiologists together
RESEARCH ARTICLE

Profiling bacterial communities in low biomass samples: pitfalls and considerations

Lisa F Stinson A C , Jeffrey A Keelan B and Matthew S Payne B
+ Author Affiliations
- Author Affiliations

A School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia

B Division of Obstetrics and Gynaecology, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, WA, Australia

C Tel: +61 6488 3200, Fax: +61 6488 7086, Email: lisa.stinson@uwa.edu.au

Microbiology Australia 40(4) 181-185 https://doi.org/10.1071/MA19053
Published: 14 November 2019

Abstract

Bacterial 16S rRNA gene sequencing studies are popular across many fields of biology. This technique has allowed us to study bacterial communities like never before, leading to significant insights into microbial ecology and host– microbe interactions. However, 16S rRNA gene-based workflows are vulnerable to confounding and bias at every step. Many studies are plagued by entrenched methodological errors, producing data riddled with experimental artefacts. These issues are amplified in the study of low bacterial biomass samples, such as forensic and ancient samples, blood, meconium, ice and the built environment. It is, therefore, necessary to define the pitfalls of low biomass 16S rRNA gene-based work flows and to identify methods that may allow more accurate characterisation of bacterial communities in such samples.


References

[1]  Karstens, L. et al. (2019) Controlling for contaminants in low-biomass 16S rRNA gene sequencing experiments. mSystems 4, e00290-19.
Controlling for contaminants in low-biomass 16S rRNA gene sequencing experiments.Crossref | GoogleScholarGoogle Scholar | 31164452PubMed |

[2]  Weyrich, L.S. et al. (2019) Laboratory contamination over time during low-biomass sample analysis. Mol. Ecol. Resour. 19, 982–996.
Laboratory contamination over time during low-biomass sample analysis.Crossref | GoogleScholarGoogle Scholar | 30887686PubMed |

[3]  Glassing, A. et al. (2016) Inherent bacterial DNA contamination of extraction and sequencing reagents may affect interpretation of microbiota in low bacterial biomass samples. Gut Pathog. 8, 24.
Inherent bacterial DNA contamination of extraction and sequencing reagents may affect interpretation of microbiota in low bacterial biomass samples.Crossref | GoogleScholarGoogle Scholar | 27239228PubMed |

[4]  Weiss, S. et al. (2014) Tracking down the sources of experimental contamination in microbiome studies. Genome Biol. 15, 564.
Tracking down the sources of experimental contamination in microbiome studies.Crossref | GoogleScholarGoogle Scholar | 25608874PubMed |

[5]  Grahn, N. et al. (2003) Identification of mixed bacterial DNA contamination in broad-range PCR amplification of 16S rDNA V1 and V3 variable regions by pyrosequencing of cloned amplicons. FEMS Microbiol. Lett. 219, 87–91.
Identification of mixed bacterial DNA contamination in broad-range PCR amplification of 16S rDNA V1 and V3 variable regions by pyrosequencing of cloned amplicons.Crossref | GoogleScholarGoogle Scholar | 12594028PubMed |

[6]  Kulakov, L.A. et al. (2002) Analysis of bacteria contaminating ultrapure water in industrial systems. Appl. Environ. Microbiol. 68, 1548–1555.
Analysis of bacteria contaminating ultrapure water in industrial systems.Crossref | GoogleScholarGoogle Scholar | 11916667PubMed |

[7]  McAlister, M.B. et al. (2002) Survival and nutritional requirements of three bacteria isolated from ultrapure water. J. Ind. Microbiol. Biotechnol. 29, 75–82.
Survival and nutritional requirements of three bacteria isolated from ultrapure water.Crossref | GoogleScholarGoogle Scholar | 12161774PubMed |

[8]  Minich, J.J. et al. (2019) Quantifying and understanding well-to-well contamination in microbiome research. mSystems 4, e00186-19.
Quantifying and understanding well-to-well contamination in microbiome research.Crossref | GoogleScholarGoogle Scholar | 31239396PubMed |

[9]  de Goffau, M.C. et al. (2019) Human placenta has no microbiome but can contain potential pathogens. Nature 572, 329–334.
Human placenta has no microbiome but can contain potential pathogens.Crossref | GoogleScholarGoogle Scholar | 31367035PubMed |

[10]  Seitz, V. et al. (2015) A new method to prevent carry-over contaminations in two-step PCR NGS library preparations. Nucleic Acids Res. 43, e135.
A new method to prevent carry-over contaminations in two-step PCR NGS library preparations.Crossref | GoogleScholarGoogle Scholar | 26152304PubMed |

[11]  Salter, S.J. et al. (2014) Reagent and laboratory contamination can critically impact sequence-based microbiome analyses. BMC Biol. 12, 87.
Reagent and laboratory contamination can critically impact sequence-based microbiome analyses.Crossref | GoogleScholarGoogle Scholar | 25387460PubMed |

[12]  Stinson, L.F. et al. (2019) Identification and removal of contaminating microbial DNA from PCR reagents: impact on low-biomass microbiome analyses. Lett. Appl. Microbiol. 68, 2–8.
Identification and removal of contaminating microbial DNA from PCR reagents: impact on low-biomass microbiome analyses.Crossref | GoogleScholarGoogle Scholar | 30383890PubMed |

[13]  Eisenhofer, R. et al. (2019) Contamination in low microbial biomass microbiome studies: issues and recommendations. Trends Microbiol. 27, 105–117.
Contamination in low microbial biomass microbiome studies: issues and recommendations.Crossref | GoogleScholarGoogle Scholar | 30497919PubMed |

[14]  de Goffau, M.C. et al. (2018) Recognizing the reagent microbiome. Nat. Microbiol. 3, 851–853.
Recognizing the reagent microbiome.Crossref | GoogleScholarGoogle Scholar | 30046175PubMed |

[15]  Stinson, L.F. et al. (2019) Characterization of the bacterial microbiome in first-pass meconium using propidium monoazide (PMA) to exclude nonviable bacterial DNA. Lett. Appl. Microbiol. 68, 378–385.
Characterization of the bacterial microbiome in first-pass meconium using propidium monoazide (PMA) to exclude nonviable bacterial DNA.Crossref | GoogleScholarGoogle Scholar | 30674082PubMed |

[16]  Emerson, J.B. et al. (2017) Schrödinger’s microbes: tools for distinguishing the living from the dead in microbial ecosystems. Microbiome 5, 86.
Schrödinger’s microbes: tools for distinguishing the living from the dead in microbial ecosystems.Crossref | GoogleScholarGoogle Scholar | 28810907PubMed |

[17]  Fittipaldi, M. et al. (2012) Progress in understanding preferential detection of live cells using viability dyes in combination with DNA amplification. J. Microbiol. Methods 91, 276–289.
Progress in understanding preferential detection of live cells using viability dyes in combination with DNA amplification.Crossref | GoogleScholarGoogle Scholar | 22940102PubMed |

[18]  Vaishampayan, P. et al. (2013) New perspectives on viable microbial communities in low-biomass cleanroom environments. ISME J. 7, 312–324.
New perspectives on viable microbial communities in low-biomass cleanroom environments.Crossref | GoogleScholarGoogle Scholar | 23051695PubMed |

[19]  Weinmaier, T. et al. (2015) A viability-linked metagenomic analysis of cleanroom environments: eukarya, prokaryotes, and viruses. Microbiome 3, 62.
A viability-linked metagenomic analysis of cleanroom environments: eukarya, prokaryotes, and viruses.Crossref | GoogleScholarGoogle Scholar | 26642878PubMed |

[20]  Checinska Sielaff, A. et al. (2019) Characterization of the total and viable bacterial and fungal communities associated with the International Space Station surfaces. Microbiome 7, 50.
Characterization of the total and viable bacterial and fungal communities associated with the International Space Station surfaces.Crossref | GoogleScholarGoogle Scholar | 30955503PubMed |

[21]  Kommedal, Ø. et al. (2012) Dual priming oligonucleotides for broad-range amplification of the bacterial 16S rRNA gene directly from human clinical specimens. J. Clin. Microbiol. 50, 1289–1294.
Dual priming oligonucleotides for broad-range amplification of the bacterial 16S rRNA gene directly from human clinical specimens.Crossref | GoogleScholarGoogle Scholar | 22278843PubMed |

[22]  Radomski, N. et al. (2013) The critical role of DNA extraction for detection of mycobacteria in tissues. PLoS One 8, e78749.
The critical role of DNA extraction for detection of mycobacteria in tissues.Crossref | GoogleScholarGoogle Scholar | 24194951PubMed |

[23]  Bachmann, N.L. et al. (2018) Advances in clinical sample preparation for identification and characterization of bacterial pathogens using metagenomics. Front. Public Health 6, 363.
Advances in clinical sample preparation for identification and characterization of bacterial pathogens using metagenomics.Crossref | GoogleScholarGoogle Scholar | 30619804PubMed |

[24]  Marotz, C.A. et al. (2018) Improving saliva shotgun metagenomics by chemical host DNA depletion. Microbiome 6, 42.
Improving saliva shotgun metagenomics by chemical host DNA depletion.Crossref | GoogleScholarGoogle Scholar | 29482639PubMed |

[25]  Doughty, E.L. et al. (2014) Culture-independent detection and characterisation of Mycobacterium tuberculosis and M. africanum in sputum samples using shotgun metagenomics on a benchtop sequencer. PeerJ 2, e585.
Culture-independent detection and characterisation of Mycobacterium tuberculosis and M. africanum in sputum samples using shotgun metagenomics on a benchtop sequencer.Crossref | GoogleScholarGoogle Scholar | 25279265PubMed |

[26]  Votintseva, A.A. et al. (2017) Same-day diagnostic and surveillance data for tuberculosis via whole-genome sequencing of direct respiratory samples. J. Clin. Microbiol. 55, 1285–1298.
Same-day diagnostic and surveillance data for tuberculosis via whole-genome sequencing of direct respiratory samples.Crossref | GoogleScholarGoogle Scholar | 28275074PubMed |

[27]  Schrader, C. et al. (2012) PCR inhibitors – occurrence, properties and removal. J. Appl. Microbiol. 113, 1014–1026.
PCR inhibitors – occurrence, properties and removal.Crossref | GoogleScholarGoogle Scholar | 22747964PubMed |

[28]  Stinson, L.F. et al. (2018) Comparison of meconium DNA extraction methods for use in microbiome studies. Front. Microbiol. 9, 270.
Comparison of meconium DNA extraction methods for use in microbiome studies.Crossref | GoogleScholarGoogle Scholar | 29515550PubMed |

[29]  Al-Soud, W.A. and Radstrom, P. (2001) Purification and characterization of PCR-inhibitory components in blood cells. J. Clin. Microbiol. 39, 485–493.
Purification and characterization of PCR-inhibitory components in blood cells.Crossref | GoogleScholarGoogle Scholar | 11158094PubMed |

[30]  Stinson, L.F. et al. (2019) The not-so-sterile womb: evidence that the human fetus is exposed to bacteria prior to birth. Front. Microbiol. 10, 1124.
The not-so-sterile womb: evidence that the human fetus is exposed to bacteria prior to birth.Crossref | GoogleScholarGoogle Scholar | 31231319PubMed |

[31]  Davis, N.M. et al. (2018) Simple statistical identification and removal of contaminant sequences in marker-gene and metagenomics data. Microbiome 6, 226.
Simple statistical identification and removal of contaminant sequences in marker-gene and metagenomics data.Crossref | GoogleScholarGoogle Scholar | 30558668PubMed |

[32]  Payne, M.S. et al. (2019) Re: ‘Amniotic fluid from healthy term pregnancies does not harbor a detectable microbial community’ (2018) 6:87, https://doi.org/10.1186/s40168-018-0475-7. Microbiome 7, 20.
Re: ‘Amniotic fluid from healthy term pregnancies does not harbor a detectable microbial community’ (2018) 6:87, https://doi.org/10.1186/s40168-018-0475-7.Crossref | https://doi.org/10.1186/s40168-018-0475-7.&journal=Microbiome&volume=7&pages=20-&publication_year=2019&author=M%2ES%2E%20Payne&hl=en&doi=10.1186/s40168-019-0642-5" target="_blank" rel="nofollow noopener noreferrer" class="reftools">GoogleScholarGoogle Scholar | 30755258PubMed |