Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Australian Dental Journal

The official journal of the Australian Dental Association


Australian Dental Journal 2018; 63: 14–24

doi: 10.1111/adj.12565

Studying the human oral microbiome: challenges and the


evolution of solutions
AML Benn,* NCK Heng,† JM Broadbent,† WM Thomson†
*Southern District Health Board, Dunedin, New Zealand.
†The University of Otago – Sir John Walsh Research Institute, Dunedin, New Zealand.

ABSTRACT
Since the pioneering work of van Leeuwenhoek in 1684, subsequently built upon by other renowned microbiologists
Robert Koch, Willoughby Miller and GV Black, oral microbiology has developed innovative techniques to study the oral
microflora (now termed the ‘oral microbiome’). The advent of molecular techniques such as DNA–DNA hybridization,
polymerase chain reaction and DNA sequencing has created an array of opportunities to construct a comprehensive pic-
ture of the diversity and composition of the oral microbiome. Approximately 700 oral bacterial species have been identi-
fied, of which 50% have yet to be cultivated, and some of these are known only by their signature DNA sequences. The
synergism of ever-evolving culture-based and state-of-the-art culture-independent molecular techniques has facilitated in-
depth understanding of the dynamics, acquisition and transfer of oral bacteria, along with their role in oral and general
health and disease. Further research is needed to not only analyse but also to make sense of the ever-increasing volumes
of data which these molecular techniques (especially high-throughput DNA sequencing) are generating, as well as why
particular bacteria are present and what they are ‘actually doing’ there. This review presents a comprehensive literature
search of oral microbiology-related methods currently used to study the oral microbiome.
Keywords: DNA–DNA hybridization, next-generation DNA sequencing, oral microbiology, oral bacteria, polymerase chain reaction.
Abbreviations and acronyms: FISH = fluorescence in situ hybridization; NGS = next-generation DNA sequencing; PCR = polymerase
chain reaction.
(Accepted for publication 23 August 2017.)

In 1684, Antonie van Leeuwenhoek stated in a let-


INTRODUCTION
ter to the Royal Society of London that ‘The number
The oral cavity is home to one of the most complex, of these Animals [bacteria] in the scurf of a man’s
dynamic and diverse microbial collections in the Teeth are so many that I believe they exceed the num-
human body. This collection, termed the ‘oral micro- ber of Men in a kingdom’.8–11 Van Leeuwenhoek
biome’, comprises mainly (eu)bacteria, viruses, fungi, examined plaque samples and observed that the type
protozoa and archaebacteria (Archaea). It is also the and numbers of different ‘Animals’ varied among indi-
oldest recognized microbial ecosystem. Current esti- viduals. His drawings were among the first to illus-
mates suggest that up to 1000 bacterial species are trate the main morphological types of bacteria – that
present in the mouth, inhabiting several distinct is, coccus, rod, vibrio and spirillum – in this complex
microbial niches. These include saliva, the teeth, the community.9–12
gingival sulcus, the attached gingiva, the tongue, the Almost two centuries later, Robert Koch’s develop-
cheek, the lip, and the hard and soft palate.1–3 Within ment, in 1881, of a solidified culture medium was a
each oral habitat, the microbes can be found growing momentous advance in microbiology. He developed a
in a distinct community, or biofilm, a functionally readily reproducible technique for growing and isolat-
and structurally organized, matrix-enclosed aggregate ing pure cultures of microorganisms using gelatin and
of microorganisms which adheres to surfaces such as eventually agar. This technique enabled researchers to
tooth enamel. That the oral microbiome is readily cultivate, identify, name and classify different
accessible and easily sampled has resulted in it being microbes,9 including many of those causing serious
the most-studied human microbiome, and it serves as infections in humans.13 Koch’s postulates for identify-
a model for biofilm biology in general.3–7 ing the causative agent for a given infectious disease,

14 © 2017 Australian Dental Association


Studying the human oral microbiome

together with his culture medium innovation, not only communities, using methods biased towards those
earned him the 1905 Nobel Prize, but also resulted in bacteria which survive transportation in a sample, can
a microbiological world divided into the ‘cultured’ grow easily or rapidly in the laboratory, and that are
and the ‘yet-to-be cultured’.14 amenable to genetic modification.20,23 Despite the
Credited as being one of the first oral microbiolo- abundance of knowledge about opportunistic patho-
gists was WD Miller, a dental scientist and Professor gens within the oral microbiota, it is limited and fun-
of Operative Dentistry who worked with Koch at the damentally incomplete. The ‘overlooked’ bacteria,
University of Berlin.11 In 1890, 9 years after Koch’s known by their molecular signatures, may be respon-
innovative developments and formulations, Miller sible for several oral and general diseases.13 Indeed,
published his seminal work ‘Micro-organisms of the without studying the several hundred uncultivable spe-
human mouth: the local and general diseases which cies found in healthy and diseased sites in the human
are caused by them’. He reported isolating more than mouth, many aspects of microbial dynamics in the
100 bacterial types of bacteria from the juices and oral cavity may never be understood.17,19,24 Hence,
deposits in the mouth.15 He also noted and high- one of the major challenges facing oral microbiology
lighted microbiological issues that are still pertinent is the ability to culture the as-yet-to-be cultivated
today, namely that numerous oral bacteria are resis- 50% of oral species.
tant to cultivation, and that the oral cavity is an open Advances in knowledge of oral microbial ecology
system, with the number and variety of bacteria con- and dynamics have enabled development of novel cul-
tinually being augmented by new microbes from air, ture techniques. The requirements for the cultivation of
food and drink.15 Concurrently, GV Black (then Pro- numerous oral bacteria species include distinct condi-
fessor of Oral Pathology at the Missouri Dental Col- tions such as an anaerobic (oxygen-free) environment,
lege) reported culturing and identifying bacteria from incubation in a variety of temperatures, chemically-
samples of carious tooth material.16 defined media containing specific amounts of nutrients,
cytokine networks and microbial co-colonizers.13,24
Despite these advances, many organisms may remain
CULTIVATION
uncultivable in the conventional manner because they
Historically, guided by Koch’s postulates, the quest exist in obligate metabolic associations with other
has been to identify and characterize cultivable organisms. In reality, oral bacteria do not live in isola-
bacteria that were associated with various oral dis- tion but in complex communities called biofilms, char-
eases. However, culture has limitations in revealing acterized by multiple growth dependencies, synergies
the actual diversity of the oral microbiome. In 1890, and antagonisms, along with mutual reliance for
Miller himself commented on his inability to culture growth and survival.3,24 In vitro cultivation possibly
all the bacteria he observed.6,16,17 This discrepancy remains elusive for many species because of a lack of
between population sizes and variety as estimated essential nutrients, growth factors and/or signalling
from microscopy and culturing is known as the ‘great molecules, overfeeding conditions, lack of cross-feeding
plate count anomaly’.14,18,19 Hence, laboratory-based partners, culture media toxicity and disruption of bac-
culture per se is unable to fully characterize the diver- terial quorum-sensing and other signalling systems.17
sity and complexity of bacterial communities such as There may be other, unknown reasons for some species
those found in the oral cavity.20 Consequently, cultur- not yet having been cultivated.
ing and functional analysis of as-yet uncultivated Conventional cultivation of bacteria samples requires
microorganisms remains a challenge in microbiological taking a sample, transferring the sample to an appropri-
research.19,21 ate medium for transportation, and storage following
It is estimated that more than 99% of bacterial collection. This is followed by dispersion and plating
species on Earth have yet to be cultured in the lab- the bacteria onto various culture media in the labora-
oratory17,20 but the endeavours of numerous tory. The bacteria are then isolated and characterized
researchers (building on the work of Miller and by their colony morphologies (appearance) and bio-
Black) have now isolated, cultivated, identified, chemical testing. Species which do not grow are natu-
characterized and classified approximately 50% of rally overlooked. Molecular techniques have enabled
the estimated 700 bacterial species which commonly an more in-depth investigation of mixed bacterial com-
colonize the human mouth.13,19,22 However, the munities. In turn, these techniques have resulted in dif-
remaining 50% of oral bacterial species identified ferent and more focused approaches to sample
through more recently-developed culture-independent cultivation. Initially, the species in the sample may be
DNA sequencing approaches remain resistant to identified by culture-independent molecular methods.
cultivation.6,13,16,17,19,22 Cultivation can then be attempted using information
Current concepts of oral disease are based largely such as the site or sites (subgingival, supragingival) and
on knowledge obtained from fragments of bacterial distinct conditions (substrate availability, co-colonizing
© 2017 Australian Dental Association 15
AML Benn et al.

species) under which the bacteria occur in the mouth. The process of categorizing bacteria phylogenetically
However, many bacteria remain resistant to cultivation is closely related to taxonomy and is the means of
despite this advance.22 classifying organisms in an ordered hierarchical sys-
Contemporary strategies for cultivation include cul- tem showing evolutionary relationships. It aims to
ture media with few or no added nutrients, long-term improve the understanding of bacterial function in
cultivation, serial dilution of slow-growing bacteria, community structures, biodiversity and environments.
addition of specific growth factors to media, and Species is the basic taxonomic group in bacterial sys-
in vivo incubation. In addition, the availability of tematics; in ascending order, each species belongs to a
sequenced genomes has created the possibility of using genus, family, class, phylum/division and domain.20,27
computer modelling of metabolic networks to aid in The oral cavity has a complex microbial ecology
the development of bespoke culture media.17 For and a rich biological setting with a number of distinc-
example, Sizova et al.19 used a combination of some tive niches, each of which provides a unique habitat
of these novel strategies and traditional cultivation for microbial colonization. These niches include the
techniques to isolate and culture 10 strains previously teeth, the gingival sulcus, the tongue, the floor of the
known only by their molecular signature, as well as mouth, the cheek, the hard and soft palates, the ton-
20 new species. Similarly, using a variety of molecular sils, the throat and the saliva.2,5,7,28 The most com-
and traditional microbiological approaches, Soro monly-used molecular research techniques used in
et al.24 and He et al.21 were able to culture and char- oral molecular microbiological research include DNA
acterize members of phylum TM7, a relatively new hybridization, polymerase chain reaction (PCR) and
phylogenetic group of bacteria previously identified by DNA sequencing. In the past, they were conducted as
next-generation DNA sequencing (NGS) and largely stand-alone techniques but, at the present time, PCR
believed to be unculturable. and DNA sequencing are invariably intertwined.
Cultivation remains the cornerstone of oral microbi- A consequence of research using culture-independent
ology in characterizing phenotypically and formally approaches was the generation of thousands of sequences
naming species, as well as describing the physiology of cloned human oral bacterial 16S rRNA genes, which
and pathogenicity of particular species. However, it is were deposited into DNA sequence databases such as
now enhanced and informed by the development and GenBank without any taxonomic reference points. In the
advances in molecular techniques, thereby enabling absence of any taxonomic (naming) scheme, researchers
and enriching the study of complex host-associated were publishing findings using isolated 16S rRNA gene
bacterial communities such as those in the oral cavity. sequences (clones) as provisional taxonomic names.
Moreover, in order to phylogenetically place an oral
clone, investigators had to manually align sequences and
MOLECULAR ORAL MICROBIOLOGY
generate their own phylogenetic trees.5
The discovery of the double helical structure of the The recognition of this problem led to researchers
DNA molecule by Nobel laureates James Watson and such as Chen and Dewhirst creating a standardized,
Francis Crick in 1953 is arguably one of the most universal taxonomic scheme, namely the Human Oral
important scientific discoveries of the 20th century.25 It Microbiome Database (HOMD; www.homd.org), a
explained the mechanism of base pairing by which curated phylogeny-based database of 16S rRNA gene
genetic information is stored and copied in living sequences. Because 16S rRNA gene sequencing is
organisms. This ground-breaking work paved the way insufficient for formal species assignment of bacteria,
for those seeking to detect, identify, type and under- the taxonomic scheme that was developed is a provi-
stand microorganisms in order to develop more sophis- sional one; the guidelines governing formal naming of
ticated culture-independent, nucleic acid-based microbial species still require isolation of a pure cul-
molecular technologies and techniques. These collec- ture and full phenotypic, and preferably genomic,
tively have revolutionized the field of oral microbiology characterization. The HOMD was the first description
because they have revealed a degree of biodiversity of a human-associated microbiome. It enables
among oral bacteria which has exceeded expectations. researchers to relate sequence information to specific
The dynamics of the oral microbiota are now better organisms in a taxonomic framework which is key to
understood because of the identification of patterns of understanding the role of the microbiome in health
acquisition and transmission of bacteria.20,26 and disease.5 To date, approximately 46% of the
In addition, the identification of universally-con- HOMD 16S rRNA gene reference sequence taxa are
served DNA sequences such as the 16S rRNA gene validly-named species, 14% are unnamed (but culti-
made information about evolutionary relatedness vated), and 32% are unnamed and uncultured taxa
among various groups of organisms (phylogenetics) known primarily from 16S rRNA sequence informa-
available. In turn, this has provided a universal system tion. The number of species residing in the human
for the identification and categorization of bacteria. oral cavity ranges from 700 to over 1000.2,5
16 © 2017 Australian Dental Association
Studying the human oral microbiome

Checkerboard DNA–DNA hybridization was first


CONTEMPORARY MOLECULAR TECHNIQUES TO
described in 1994 by Socransky et al.32 In this tech-
PROFILE THE ORAL MICROBIOTA
nique, DNA standards representing the target species
Culture independent nucleic acid-based molecular are immobilised on a nylon membrane in an array
analysis techniques are used for detecting and identify- format, then simultaneously cross-hybridized with
ing individual or multiple bacteria, bacterial commu- (usually) radioactively-labelled genomic DNA probes.
nity diversity and bacterial typing.20 Essentially, However, cultivable bacteria are needed in order to
molecular techniques need a macromolecule of inter- provide the genomic DNA to construct the probes.
est (DNA, RNA or protein), method-specific reagents, Checkerboard DNA–DNA hybridization provides use-
automated sample handling such as those handled by ful quantitative data and is useful in the study of the
robots and, ideally, a bioinformatics platform which complex biodiverse plaque microbiota in both small-
allows computer-aided analyses of large molecular and large-scale studies. Initially used in the study of
datasets to be carried out.29 The molecular techniques periodontal disease, the technique has since been
routinely used in oral microbiology are largely DNA- employed to study the oral microbiota in diverse areas
based and can be divided fundamentally into three such as microbial ecology, general health and disease,
broad categories, namely: (i) DNA hybridization; (ii) smoking, cariology and endodontics.33–40
PCR; and (iii) DNA sequencing. The techniques avail- Reverse-capture oligonucleotide hybridization is a
able today are essentially more advanced high- modification of the checkerboard method involving
throughput approaches which incorporate combina- PCR amplification of the sample’s 16S rRNA and spe-
tions of the three categories. cies-specific 16S rRNA probes. These probes can be
constructed to identify both cultivable and uncul-
tivable bacteria.41 In 2006, Paster et al.42 introduced
DNA–DNA hybridization
a further modification using a microarray format
The discovery of the hybridization reaction, which (glass slides) instead of a nylon membrane. This
is the spontaneous pairing of two complementary method, together with visualization scanners and
(i.e. matching sequences) strands of a nucleic acid advanced computer-based analysis, can detect a large
double-helix, led to the development of DNA– number of species simultaneously.20,30,44 This even-
DNA hybridization techniques for the study of tually led to the development of the sophisticated
microorganisms. These techniques utilize single- human oral microbe identification microarray
stranded ‘DNA probes’ labelled with either a which allows the detection of approximately 300
radioactive isotope, fluorescent or chemiluminescent bacterial species.20,43,44 Reverse-capture oligonu-
tag that bind to complementary bases of a target cleotide hybridization and modifications of the tech-
DNA strand to form a duplex and can then be nique have been used in researching topics such as
detected using an appropriate instrument or combi- dental caries, endodontics, oral cancer, HIV and
nation of chemical reagents. The probes used in periodontitis.20,42,43,45
the annealing process are either whole-genome The FISH method, first introduced in 1980, makes
DNA probes or short (15–30-base) oligonucleotide use of the whole cell.46 Fluorescently-labelled oligonu-
probes with DNA sequences which complement cleotides probes designed to target the rRNA gene are
their target sequence.20,30 hybridized to partially-fixed, whole cells. The probes
Whole-genome probes are more likely to cross-react can then be directly visualized using fluorescent
with non-target bacteria than the more specific microscopy or combined with flow cytometry for
oligonucleotide probes such as those binding to analysis of biodiverse microbial samples.30,43 FISH
regions of the 16S rRNA gene. The 16S rRNA gene can be used for detection of cultivable and as-yet
enables precise taxonomic positioning of bacteria uncultivated bacteria, in the study of morphology,
because, in species with 70% or more genomic simi- taxonomic identification, quantity, spatial organiza-
larity, the 16S rRNA gene is usually a more than tion and biofilm architecture.12,20,30,43,47 The applica-
97% gene sequence match; this is the universally- tion of FISH in oral microbiological research has been
accepted cut-off for species assignment.20,30,31 in fields such as biofilm structure, genome–genome
Hybridization techniques used in oral microbiological interactions, subgingival organisms and endodon-
research include whole-genome checkerboard DNA– tics.12,30,47 As with all of the molecular methods,
DNA hybridization, reverse-capture oligonucleotide FISH has continued to evolve. For example, it can
hybridization (modification of the checkerboard now be used to investigate the system-level taxonomic
method), fluorescence in situ hybridization (FISH) and spatial structure of complex biofilms such as plaque
DNA microarray technology. Table 1 presents a sum- because of the development of combinational labelling
mary of the principles, applications, advantages and and spectral imaging FISH, which expanded the num-
limitations of these methods. ber of different taxa distinguishable in a single field.12
© 2017 Australian Dental Association 17
AML Benn et al.

Table 1. DNA hybridization-based techniques


Technique Principle Application Advantages Limitations

Whole-genomic Hybridization of a selection of Screening for selected Rapid, direct, sensitive, Lower specificity than
checkerboard DNA radioactively- or dye-labelled bacteria relatively inexpensive oligonucleotide probes
–DNA whole genomic DNA probes to Enumeration of species in analysis of large number
hybridization sets of sample DNA fixed on a complex systems of samples for high level Requires cultivable
membrane of different bacteria bacteria
Reverse-capture Hybridization of a selection of Screening for selected Highly selective under Low sensitivity when
oligonucleotide labelled polymerase chain bacteria stringent conditions low levels of target
hybridization reaction (PCR)-amplified 16S bacteria in complex
rRNA gene segments from sets Precise hybridization of samples with bacterial
of samples to species-specific complementary target and human DNA
oligonucleotide probes fixed on sequences
a membrane. This is a Loss of quantitative
Precise taxonomy assessment
modification of the checkerboard
technique
Fluorescence in situ In situ hybridization of Detection, direct Identification and Labour-intensive
hybridization fluorescent-labelled 16S rRNA visualization and quantification of several
(FISH) oligonucleotide probes to identification of single species in same sample Requires 16S rRNA
bacterial call rRNA in the microbial cells gene sequence data
sample Spatial organization and Small number species
detection when used with distinguishable in a
confocal laser scanning sample
microscopy
DNA microarray Hybridization of labelled DNA Detection and Wide applicability Expensive
sequences in the sample to identification of
target-specific oligonucleotides pathogens and bacteria in Vast amount of Hybridization
fixed onto a nylon mixed infections or information specificity
(polyvinylidene difluoride) contaminations Simultaneous analysis of Quantification of
membrane or a glass slide extensive number of signals – noise ratios
different bacteria Lack of probes for
unknown bacteria
Detection of target
bacteria present at
low levels
12,20,30,36
Adapted from references .

Methods using PCR thermocycling apparatus. Depending on the extension


period, DNA fragments of more than 20 kbp in
In 1985, Kary Mullis invented a process for the
length can be generated. Because amplification of the
amplification of specific DNA fragments from any
target DNA fragments is exponential, minute amounts
DNA molecule. This process, called the PCR, eventu-
of DNA can be multiplied billions of times in a matter
ally earned Mullis the Nobel Prize in 1993. Since its
of hours. PCR is extremely sensitive and highly spe-
inception, PCR has become a central technique in a
cific.30,48–51 However, there are limitations, particu-
myriad of molecular techniques. It can be used either
larly within heterogeneous bacterial samples such as
as a stand-alone method or as the first step in generat-
oral biofilms. These include: (i) errors from the need
ing starting material for other molecular techniques
for different methods for lysis of gram-negative and
such as DNA sequencing. The basic PCR technique,
gram-positive bacteria; (ii) oral samples containing
as practised today, relies on two specific oligonu-
amplification inhibitors such as blood; (iii) DNA
cleotide primers (usually 20–30 nucleotides long) that
sequence differences leading to unequal DNA denatu-
frame the target DNA sequence, namely the template,
ration and annealing causing amplification bias; and
which is then amplified by a thermostable DNA poly-
(iv) erroneous findings from the amplification of con-
merase enzyme. The dsDNA template is first heat-
taminating DNA.20 In addition, PCR is potentially
denatured (>94 °C) and then quickly cooled to 50–55
time-consuming for complex biodiverse systems
°C to allow the primers to bind (anneal) to the tem-
because primers and amplification conditions may
plate. After a period of annealing (usually 30–60 s),
have to be optimized for each target fragment.52
the temperature is raised to 68–72 °C to allow the
Pertinent information relating to conventional PCR
DNA polymerase to synthesize new DNA strands
and its derivatives is presented in Table 2. Conven-
(also known as extension). Each newly-synthesized
tional PCR and its many variants have been used in
DNA strand then acts as a new template in the next
oral microbiology research, examples of which include
cycle. The cycles of denaturation, annealing and
periodontal health and disease, epidemiology, cariol-
extension are then repeated up to 40 times in a
ogy, genotype diversity, micro-ecology, endodontics,
18 © 2017 Australian Dental Association
Studying the human oral microbiome

Table 2. Polymerase chain reaction (PCR)-based methods


Method Principle Application Limitations Advantages

Conventional PCR Amplification of a single Detection and Formation of non-specific Extremely small sample can
(PCR) gene region using two identification of bacteria products be used
oligonucleotide primers Amount of amplified Identification of cultivable
(multiplex PCR amplifies product correlates poorly and non-cultivable bacteria
several genetic regions in with original DNA
a single reaction) template amount
Real-time quantitative Quantitative monitoring of Single species detection in Laborious and expensive PCR product monitoring as
PCR (q-PCR, qRT- the information of PCR epidemiological studies in early stages of the reaction happens
PCR) product in real time by development Closed system reduces risk
detection of incorporated May underestimate species of amplicons release into
fluorescent dyes of diversity in a community laboratory
specific wavelengths sample Provides quantitative data
PCR-denaturing Electrophoretic separation Studying bacterial diversity No phylogenetic Fast
gradient of PCR-amplified 16S (detection and identification Semi-quantitative
gel electrophoresis rRNA gene fragments identification of all Bias introduced by PCR
(PCR-DGGE) through a polyacrylamide bacteria)
gel containing an Monitor community
increasing gradient of dynamics
denaturants Identify predominant
species in a community
Random amplified PCR amplification of Distinguishing condition/ Poor reproducibility of No previous sequence DNA
polymorphic DNA/ genomic DNA using a disease-associated species band patterns information required
arbitrarily primed single random primer Easy to use
PCR (RAPD/AP-PCR) under low-stringency Primer annealing non-
conditions and stringent
subsequent size separation Enables annealing to
by agarose gel unknown DNA sequences
electrophoresis of low complementarity
Repetitive element- PCR amplification using Genotype profile analysis Reproducibility may be High discriminatory power
based PCR (Rep-PCR) consensus primers for unreliable Inexpensive
randomly dispersed
palindromic sequences in
genomic DNA and size
separation by agarose gel
electrophoresis
Multilocus sequence Comparison of sequences Distinguishing strains of a Expensive High discriminatory power
typing (MLST) of PCR amplified known species Low throughput Portable
housekeeping gene Standard nomenclature
between test and No ambiguity of nucleotide
reference strains sequencing
PCR-restriction Restriction digestion of the Genotyping Mass of gel bands Requires small amount of
fragment PCR amplified target and resulting in difficulty in sample DNA
length polymorphism fragment size separation comparing strains Fast
(PCR-RFLP) by gel electrophoresis No phylogenetic Semi-quantitative
identification Inexpensive
PCR bias Prior testing using sequence
data of restriction enzyme
discrimination capacity
and restriction fragment
sizes
Terminal restriction Restriction digestion of Community profiling Underestimate species Provides knowledge of
fragment length PCR amplified 16S rDNA Monitoring changes over diversity in community taxonomic position of test
polymorphism (T- using fluorescent-labelled time or after known sample bacteria
RFLP) forward primer and alterations
detection by DNA
sequencer
20,26,39,64–67
Adapted from references .

bacterial communities (subgingival, supragingival and decades.58–60 The prototype method was expensive,
saliva) and antimicrobial susceptibility.20,39,53–57 labour-intensive and potentially dangerous. It
involved the use of a heat-labile DNA polymerase
enzyme, radioactively-labelled chain terminators and
DNA SEQUENCING: THE NEXT GENERATION
X-ray films. ‘Reading’ a DNA sequence had to be
In 1977, Sanger and Gilbert developed a DNA done visually. Over the decades, the Sanger technique
sequencing method (commonly referred to as ‘Sanger has been extensively improved, automated and minia-
sequencing’) that has been the gold standard for four turized by combining a PCR-based step for the DNA
© 2017 Australian Dental Association 19
AML Benn et al.

synthesis and chain termination (with dye-labelled summarised in Table 3. At the time of writing, the
dideoxynucleotides) reactions, capillary electrophore- 454 pyrosequencing and SOLiD platforms have since
sis, detection of the fluorescent dyes, and computer- been discontinued due to their non-competitive cost-
aided base-calling into a single bench top apparatus. effectiveness.
Even the all-important purification of the DNA tem- Their truly culture-independent nature means that
plate to be sequenced can be automated by using NGS systems enable hypothesis-driven studies on
robotic instruments.60,61 The first bacterial genome previously unknown and unclassified microorgan-
sequence, that of the pathogen Haemophilus influen- isms.23 These methods result in considerably greater
zae, was completed in 1995 using Sanger sequenc- depth (number of sequences per sample) and breadth
ing.59 In the early decades of molecular oral (number of samples/individuals analysed) than San-
microbiology, Sanger sequencing was used in combi- ger sequencing. NGS technology offers much lower
nation with PCR, sometimes accompanied by cloning, costs, avoids cloning biases and offers the possibility
to study the microbiota associated with various oral of bacterial genome or profiling analyses in a matter
sites, in health and in association with disease condi- of hours or days rather than months or years.23,59,61
tions such as dental caries, periodontal diseases, However, it suffers from sequencing errors, referred
endodontic infections, peri-implantitis and halitosis.61 to as ‘sequencing noise’, which can result in either
However, the main limitation of these techniques, overestimation of sample diversity or, after removal
especially when investigating the microbial composi- of the errors/sequencing noise, underestimation of
tion of complex biofilms, was the requirement for sample diversity. Researchers also have to consider
cloning genes of interest in the laboratory workhorse the compromise between generating huge amounts
Escherichia coli. This created ‘cloning bias’, whereby of data (billions of sequence reads) but with short
not all genes could be cloned due to incompatibilities read lengths (100–800 bp) and longer read lengths
with the E. coli host. (up to 30 000 bp) but with significantly fewer (sev-
In the last decade, advances in miniaturization and eral million) sequence reads. The shorter read
ingenious molecular detection methodologies have lengths limit the identification of bacterial species
resulted in an unprecedented range of so-called NGS because reliable identification of new species and
technologies. These technologies are based on either some known species ideally requires sequencing of
the DNA-by-synthesis approach – that is, still based the entire 16S rRNA gene, which is approximately
on the Sanger principle but incorporating PCR – or 1550 bp long.59 This limitation, however, may even-
more recently, the detection of changes in DNA prop- tually be circumvented by future higher-throughput
erties during the sequencing reaction. One of the first versions of ultra-long-length sequencing systems such
NGS technologies was pyrosequencing (also known as as SMRT and MinION.62,63 Moreover, technologies
‘454’), which featured an etched glass array contain- such as SMRT and MinION facilitate the sequenc-
ing over 1 000 000 individual nanolitre ‘PCR vessels’, ing of whole microbial genomes (i.e. bona fide
each capable of effecting a single PCR-based sequenc- metagenomics) within a given sample of interest,
ing reaction.60,61 The basis of pyrosequencing was thus potentially yielding much more meaningful data
detection of light upon incorporation of a fluores- on not only what microbes are present but also on
cently-labelled nucleotide. Computer software bundled their genomic secrets (e.g. metabolic pathways, viru-
with the 454 instrument (the prototype GS20 fol- lence factors).
lowed by the GS-FLX and GS-FLX+) would analyse To date, investigations into the bacteria of the oral
and provide the user with the DNA sequence reads, cavity and its various unique niches using NGS have
each averaging up to 450 bp. Variants of this princi- been wide-ranging, but they are beyond the scope of
ple with different detection schemes emerged, includ- this review. Reported in the literature have been
ing the Illumina HiSeq/MiSeq, SOLiD and research topics as diverse as the oral microbiome in
semiconductor (Ion TorrentTM) sequencing. For exam- health and disease (paediatric, adult and geriatric),
ple, Ion Torrent sequencing detects base incorporation dental plaque, supra- and subgingival bacteria, peri-
by measuring the subtle change in pH due to the odontal health and disease, endodontic infections,
release of a proton when a base is added. More microbiota of root canals (pre- and post-treatment),
recently, single DNA molecule (Pacific Biosciences cariology, oral cancer, microbiota of saliva and sys-
Single Molecule Real-Time [SMRT])62 and MinION63 temic diseases associated with oral bacteria.44,55,61
sequencing have emerged as potentially cost-effective Without a doubt, as NGS technologies are further
DNA sequencing alternatives due to their very long improved and refined to (i) be more cost- and time-
and reliable sequence read lengths (>5000 to efficient, (ii) generate fewer amplification biases and
30 000 bp).62,63 The chemistry, advantages and limi- sequencing noise, and (iii) capture data in real-time,
tations of Sanger sequencing and the most commonly- this will open up endless possibilities for further oral
used NGS sequencing technologies available today are microbiological research.23,59
20 © 2017 Australian Dental Association
Studying the human oral microbiome

Table 3. The evolution of DNA sequencing


Technique Year of Chemistry Advantages Limitations
introduction

First-generation sequencing
Sanger (dideoxy) 1977 Dye-labelled Gold standard for the last three decades Expensive ($5–10 per sequence read)
sequencing dideoxynucleotide Validation of next-generation sequencing Laborious
chain terminators results Cloning bias
Long contiguous DNA sequence reads Fairly low throughput
(up to 900 bp of reliable sequence data) Scalability
Fairly quick run times (3 hours) Speed
Resolution
Next-generation Open-ended view on whole breadth of Short length read
sequencing (NGS) microbiome Low taxonomic resolution (most to
Increase depth and sensitivity of bacterial genus level)
profile DNA and amplification bias
Enables rapid sequencing of large Sequencing errors
stretches of DNA spanning entire
genomes
Pioneer or discontinued NGS platforms
Roche/454 GS 2005 Pyrosequencing Can generate 1.2 million DNA sequences Costlier than the Illumina systems
(600 million base pairs) Relatively high error rates (>5%)
Long read length (~450 base pairs) Repetitive sequences can be its
Relatively high speed Achilles heel
Excellent for genome sequencing
Assembling scaffold for metagenomics
Life Technologies 2006 Sequencing by Very high (>99%) accuracy Short read length (50 bp)
SOLiD ligation Very high yields (60 billion base pairs) Long run times (7–14 days)
Colourspace mapping of DNA
sequence
Currently-available NGS platforms
Illumina MiSeq/ 2006 Reversible Very established system with majority Relatively short length read (up to
HiSeq terminators market share 300 bp)
Very high yield (up to 15 billion base
pairs)
High accuracy (>95%)
Reduced hands-on time
Short run times (56 h)
Lowest reagent cost
Life Technologies 2010 Change in pH upon Intermediate yields (relative to Illumina Relatively short read lengths (200–
PGM base incorporation and SMRT) of up to 1 billion base pairs 400 bp)
(Ion Torrent) Up to 5 million sequences can be Error rates of 5%
generated Prone to base homopolymer runs,
Very high accuracy (>99%) which could lead to misassemblies
Fast run times (<8 hours)
Single-molecule 2009 Dye-modified Fast cycle time High raw error rates (>5%)
real-time nucleotides Very long reads (up to >30 000 base Low throughput (hundreds of
sequencing pairs) thousands of sequences)
(SMRT) Simple preparation Highly-specialised sensitive
Low reagent costs instrument
Ideal for whole-genome sequencing and Difficult set-up
metagenomics
MinION 2008 Change in current Read length (>5 kb pairs) Relatively low throughput
(Nanopore upon base Portability – essentially ‘sequencing on a High translocation velocity
sequencing) incorporation USB stick’ Lack of nucleotide specificity
Speed
No fluorescent modification of bases
results in reduced costs and biases
Ideal for sequencing in the field (e.g.
infectious disease outbreaks)
20,59,61–63,68
Adapted from references .

Coping with DNA sequence data overload: enter the enough to generate up to millions or billions of indi-
bioinformatic tools vidual DNA sequences (Table 3), ‘denoising’ and
ensuring the integrity of these gigantic sequence data-
Up until a decade ago, DNA sequence data genera-
sets can be done only by using sophisticated and pow-
tion, despite being automated with output as readable
erful computing resources. What used to be
text files, still required verification by the human
achievable with a desktop computer now requires
researcher or teams of bioinformaticians. With the
massively-parallel multi-processing computing arrays
advent of NGS technologies that are now powerful
© 2017 Australian Dental Association 21
AML Benn et al.

comprising tens or hundreds of processing units to do information about species of interest in cases of dis-
it in the same time. These super computing arrays, ease cases such as severe early childhood caries. Our
however, would be ineffective if there were not the own research utilizing the 454 and Ion Torrent plat-
software packages to analyse the data and generate forms has revealed the presence of new genera such as
results that would be meaningful to the researchers. Granulicatella, Leptotrichia, Rothia, Veillonella and
Many software packages have been developed over phylum TM7, some of which remain recalcitrant to
the years that aim to denoise sequence data, classify conventional culture methods.73,74 Some genera
16 rRNA gene sequences into species, and assemble appear to exhibit site specificity; for example, Gran-
whole genome sequences from single-species or ulicatella can be detected in buccal swabs but not in
metagenomic data. One of the most common software dental plaque samples.73 Interestingly, S. wiggsiae has
suites used to profile bacterial communities from a not been detected in oral samples of New Zealand
variety of environments is QIIME (Quantitative children with severe dental caries, and, in some cases,
Insights Into Molecular Ecology),69 which is regularly Streptococcus sanguinis (and not S. mutans) has been
updated, and offers many types of analyses from basic found to be the prevalent species in caries lesions.73,74
species assignment to the measurement of diversity Furthermore, we observed relatively high levels of
(such as the Chao1 and Shannon diversity indices). Neisseria bacilliformis and Neisseria flavescens in
Another software package is Galaxy,70 a cloud-based severe dental caries samples, an observation
platform which features workflow options from previously unreported but which remains to be repli-
denoising through to diversity analyses. Post-analysis cated elsewhere.73,74 Similarly, our own 454-based
software may include packages such as Bioconductor NGS-based bacterial profiling of severe periodontitis
and those allowing principal component analysis. As cases has shown that Prevotella spp. (an Orange
NGS technologies have evolved to become more Complex genus), rather than P. gingivalis, was
affordable and mainstream, the composition of groups frequently detected in diseased samples.73 Our
undertaking oral microbiological research has evolved findings, albeit from relatively small samples, are
from small specialist teams to larger multidisciplinary nevertheless supported by reports from other
groups. groups.7,23,30,42,45,53,56,75,76 Current molecular oral
microbiological strategies, therefore, have evolved to
the point where we have to discard completely the
Impact of molecular microbiology on the oral
notion that oral diseases are caused by a single patho-
pathogen ‘rogues gallery’
gen (or groups of pathogens) in favour of a concept
For several decades now, students of dentistry and of a ‘healthy’ versus ‘disease-associated’ oral micro-
other oral health-related professions have been taught biome. Using NGS systems such as SMRT, which is
that the two most prevalent oral diseases in the world, ideal for metagenomic analyses, it is only a matter of
dental caries and periodontitis, are caused by the time before the roles of oral microbes in health and
gram-positive Streptococcus mutans and the gram- disease will be elucidated.
negative Porphyromonas gingivalis, respectively. This
was largely due to the ability to culture these organ-
CONCLUSION
isms using blood agar media and to these species
being amenable to genetic modification. To this day, Since van Leeuwenhoek’s initial discoveries, the fun-
much research is still being carried out on the viru- damental microbiological question remains ‘what is
lence of these organisms. With the use of checker- there?’. Most often, the answer has been simplistic
board DNA hybridization, Socransky categorized and rather opportunistic, resulting in as-yet-to-be cul-
periodontal pathogens into complexes with P. gingi- tivated bacteria and those that are present in low
valis being in the Red Complex and organisms numbers being overlooked or ignored. Molecular
exhibiting lower pathogenicity, such as Aggregatibac- methods have yielded a more comprehensive picture
ter actinomycetemcomitans, in the Orange and Yellow of composition and diversity of the oral microbiota.
Complexes.32,36 However, these methods have also resulted in a deluge
More recently, the use of 16S rRNA gene sequenc- of data, leading to the need to develop increasingly
ing, either as cloned amplicons or analysed by NGS, sophisticated computational and statistical tools in
has allowed the identification of new genera and new order to analyse and make sense of it.
species, such as Scardovia wiggsiae, in dental caries Further research is needed to reveal why particular
lesions.71,72 NGS, due to its superlative sequence bacteria are present and, more importantly, what in
yields, can also detect rare species (those representing fact they are ‘actually doing there’. Understanding
<0.001% of the microbial population in a given how bacteria interact with each other and their envi-
sample). Subsequent analyses using packages such as ronment in an open system such as the oral cavity is
QIIME and Galaxy can provide quantitative key to the ultimate aim of unravelling the complexity
22 © 2017 Australian Dental Association
Studying the human oral microbiome

of the oral microbiome and to providing a better basis 22. Leys EJ, Griffen AL, Beall C, Maiden MF. Isolation, classifica-
tion, and identification of oral microorganisms. In: Lamont RJ,
for understanding the important role oral bacteria Hajishengallis GN, Jenkinson HF, eds. Oral microbiology and
play in both oral and general health and disease. immunology. Washington: ASM Press, 2013: 77–96.
23. Zaura E. Next-generation sequencing approaches to under-
standing the oral microbiome. Adv Dent Res 2012;24:81–85.
REFERENCES
24. Soro V, Dutton LC, Sprague SV, et al. Axenic culture of a can-
1. ten Cate JM. Biofilms, a new approach to the microbiology of didate division TM7 bacterium from the human oral cavity and
dental plaque. Odontol 2006;94:1–9. biofilm interactions with other oral bacteria. Appl Environ
2. Dewhirst FE, Chen T, Izard J, et al. The human oral micro- Microbiol 2014;80:6480–6489.
biome. J Bacteriol 2010;192:5002–5017. 25. Watson JD, Crick FH. Molecular structure of nucleic acids.
3. Jenkinson HF. Beyond the oral microbiome. Environ Microbiol Nature 1953;171:737–738.
2011;13:3077–3087. 26. Kilian M. Population genetics of oral bacteria. In: Lamont RJ,
4. Foster JS, Palmer RJ, Kolenbrander PE. Human oral cavity as a Hajishengallis GN, Jenkinson HF, eds. Oral microbiology and
model for the study of genome-genome interactions. Biol Bull immunology. Washington: ASM Press, 2013: 195–208.
2003;204:200–204. 27. Haake SK, LeBlanc DJ, Tribble GD. Genetics and molecular
5. Chen T, Yu WH, Izard J, Baranova OV, Lakshmanan A, biology of oral microorganisms. In: Lamont RJ, Hajishengallis
Dewhirst FE. The Human Oral Microbiome Database: a web GN, Jenkinson HF, eds. Oral microbiology and immunology.
accessible resource for investigating oral microbe taxonomic Washington: ASM Press, 2013: 139–176.
and genomic information. Database (Oxford) 2010;2010: 28. Scannapieco FA. The oral environment. In: Lamont RJ,
baq013. Hajishengallis GN, Jenkinson HF, eds. Oral microbiology and
6. Wade WG. The oral microbiome in health and disease. Pharma- immunology. Washington: ASM Press, 2013: 51–76.
col Res 2013;69:137–143. 29. Mardis ER. A decade’s perspective on DNA sequencing tech-
7. Zaura E, Koopman JE, y Mostajo MF, Crielaard W. The Oral nology. Nature 2011;470:198–203.
Microbiome. In: JR Marchesi, ed. The human microbiota and oßcas IN. The oral microbiota: general overview,
30. Siqueira JF, R^
microbiome. Wallingford: CAB International, 2014:20–31. taxonomy, and nucleic acid techniques. In: Seymour GJ, Culli-
8. Weller CV. Antony van Leeuwenhoek: 1632-1723. Ann Internal nan MP, Heng NCK, eds. Oral biology: molecular techniques
Med 1932;6:573–584. and applications. New York: Springer, 2010: 55–69.

9. Brock TD. Milestones in microbiology. Acad Med 31. Rastogi G, Sani RK. Molecular techniques to assess microbial
1961;36:847. community structure, function, and dynamics in the environ-
ment. In: Ahmad I, Ahmad F, Pichtel J, eds. Microbes and
10. Porter JR. Antony van Leeuwenhoek: tercentenary of his dis- microbial technology: agricultural and environmental applica-
covery of bacteria. Bacteriol Rev 1976;40:260–269. tions. New York: Springer, 2011: 29–57.
11. He X-S, Shi W-Y. Oral microbiology: past, present and future. 32. Socransky S, Smith C, Martin L, Paster B, Dewhirst F, Levin A.
Int J Oral Sci 2009;1:47. Checkerboard. DNA-DNA hybridization. Biotechniques
12. Valm AM, Welch JLM, Rieken CW, et al. Systems-level analy- 1994;17:788–792.
sis of microbial community organization through combinatorial oßcas IN, Souto R, de Uzeda M, Colombo AP.
33. Siqueira JF, R^
labeling and spectral imaging. Proc Natl Acad Sci USA Checkerboard DNA-DNA hybridization analysis of endodontic
2011;108:4152–4157. infections. Oral Surg Oral Med Oral Pathol Oral Radiol
13. Wade W. Unculturable bacteria—the uncharacterized organisms Endodontol 2000;89:744–748.
that cause oral infections. J R Soc Med 2002;95:81–83. 34. Haffajee A, Socransky S. Relationship of cigarette smoking to
14. Handelsman J. Metagenomics: application of genomics to the subgingival microbiota. J Clin Periodontol 2001;28:377–
uncultured microorganisms. Microbiol Mol Biol Rev 388.
2004;68:669–685. 35. Wall-Manning G, Sissons C, Anderson S, Lee M. Checkerboard
15. Miller WD. The micro-organisms of the human mouth: the DNA–DNA hybridisation technology focused on the analysis of
local and general diseases which are caused by them. S. Karger. Gram-positive cariogenic bacteria. J Microbiol Methods
Philadelphia: S. S. White Dental Manufacturing Company 2002;51:301–311.
1890. 36. Socransky SS, Haffajee AD, Smith C, et al. Use of checkerboard
16. Black GV. Micro-organisms of the oral cavity. Ill State Dent DNA–DNA hybridization to study complex microbial ecosys-
Soc Trans 1886;22:180–208. tems. Oral Microbiol Immunol 2004;19:352–362.
oßcas IN. As-yet-uncultivated oral bacteria:
17. Siqueira JFJr, R^ 37. Li J, Helmerhorst E, Leone C, et al. Identification of early
breadth and association with oral and extra-oral diseases. J microbial colonizers in human dental biofilm. J Appl Microbiol
Oral Microbiol 2013;5:1–14. 2004;97:1311–1318.
18. Staley JT, Konopka A. Measurement of in situ activities of non- 38. Sakamoto M, Umeda M, Benno Y. Molecular analysis of
photosynthetic microorganisms in aquatic and terrestrial habi- human oral microbiota. J Periodontal Res 2005;40:277–
tats. Ann Rev Microbiol 1985;39:321–346. 285.
19. Sizova M, Hohmann T, Hazen A, et al. New approaches for 39. Haffajee AD, Yaskell T, Torresyap G, Teles R, Socransky SS.
isolation of previously uncultivated oral bacteria. Appl Environ Comparison between polymerase chain reaction-based and
Microbiol 2012;78:194–203. checkerboard DNA hybridization techniques for microbial
assessment of subgingival plaque samples. J Clin Periodontol
20. Asikainen S, Karched M, Rogers A. Molecular techniques in 2009;36:642–649.
oral microbial taxonomy identifications and typing. Norfolk,
UK: Caister Academic Press, 2008. 40. Colombo APV, Magalh~aes CB. Hartenbach FARR, do Souto
RM, da Silva-Boghossian CM. Periodontal-disease-associated
21. He X, McLean JS, Edlund A, et al. Cultivation of a human-asso- biofilm: A reservoir for pathogens of medical importance.
ciated TM7 phylotype reveals a reduced genome and epibiotic Microb Pathog 2016;94:27–34.
parasitic lifestyle. Proc Natl Acad Sci USA 2015;112:244–249.

© 2017 Australian Dental Association 23


AML Benn et al.

41. Paster BJ, Bartoszyk IM, Dewhirst FE. Identification of oral 61. Siqueira JF, Fouad AF, R^oßcas IN. Pyrosequencing as a tool for
streptococci using PCR-based, reverse-capture, checkerboard better understanding of human microbiomes. J Oral Microbiol
hybridization. Methods Cell Sci 1998;20:223–231. 2012;4:1–15.
42. Paster BJ, Olsen I, Aas JA, Dewhirst FE. The breadth of bacte- 62. Rothberg JM, Hinz W, Rearick TM, et al. An integrated semi-
rial diversity in the human periodontal pocket and other oral conductor device enabling non-optical genome sequencing. Nat-
sites. Periodontol 2000;2006(42):80–87. ure 2011;475:348–352.
43. Paster BJ, Dewhirst FE. Molecular microbial diagnosis. Peri- 63. Schloss PD, Jenior ML, Koumpouras CC, Westcott SL, High-
odontol 2000;2009(51):38–44. lander SK. Sequencing 16S rRNA gene fragments using the Pac-
44. Do T, Devine D, Marsh PD. Oral biofilms: molecular analysis, Bio SMRT DNA sequencing system. PeerJ 2016;4:e1869.
challenges, and future prospects in dental diagnostics. Clin Cos- 64. Jain M, Fiddes IT, Miga KH, Olsen HE, Paten B, Akeson M.
met Invest Dent 2013;5:11–19. Improved data analysis for the MinION nanopore sequencer.
45. Colombo APV, Boches SK, Cotton SL, et al. Comparisons of Nat Methods 2015;12:351–356.
subgingival microbial profiles of refractory periodontitis, severe 65. Fraher MH, O’Toole PW, Quigley EM. Techniques used to
periodontitis, and periodontal health using the human oral characterize the gut microbiota: a guide for the clinician. Nat-
microbe identification microarray. J Periodontol 2009;80:1421– ure Rev Gastroenterol Hepatol 2012;9:312–322.
1432. 66. Sabat AJ, Budimir A, Nashev D, et al. Overview of molecular
46. Levsky JM, Singer RH. Fluorescence in situ hybridization: past, typing methods for outbreak detection and epidemiological
present and future. J Cell Sci 2003;116:2833–2838. surveillance. Euro Surveill 2013;18:20380.
47. Kolenbrander PE, Egland PG, Diaz PI, Palmer RJ. Genome– 67. Szabo J. Molecular methods in epidemiology of methicillin
genome interactions: bacterial communities in initial dental pla- resistant Staphylococcus aureus (MRSA): advantages, disadvan-
que. Trends Microbiol 2005;13:11–15. tages of different techniques. J Med Microbiol Diagn
48. Mullis KB. The unusual origins of the polymerase chain reac- 2014;3:147.
tion. Sci Am 1990;262:56–65. 68. Liu L, Li Y, Li S, et al. Comparison of next-generation sequenc-
49. Socransky SS, Haffajee AD. Periodontal microbial ecology. Peri- ing systems. Biomed Res Int 2012;2012:1–11.
odontol 2000;2005(38):135–187. 69. Caporaso JG, Kuczynski J, Stombaugh J, et al. QIIME allows
50. Fletcher HM, Aruni W, Dou Y, Progulske-Fox A. Applied analysis of high-throughput community sequencing data. Nat-
molecular biology and the oral microbes. In: Lamont RJ, ure Meth 2010;. https://doi.org/10.1038/nmeth.f.303.
Hajishengallis GN, Jenkinson HF, eds. Oral microbiology and 70. Afgan E, Baker D, van den Beek M, et al. The Galaxy platform
immunology. Washington: ASM Press, 2013: 177–194. for accessible, reproducible and collaborative biomedical analy-
51. Nobelprize.org. Nobel Media AB 2014. URL:http://www.nobel ses: 2016 update. Nucleic Acids Res 2016;44:W3–W10.
prize.org/nobel_prizes/chemistry/laureates/1993/mullis-facts. 71. Tanner AC, Mathney JM, Kent RL, et al. Cultivable anaerobic
html. Accessed September 2016. microbiota of severe early childhood caries. J Clin Microbiol
52. Zoetendal EG, Mackie RI. Molecular methods in microbial 2011;49:1464–1474.
ecology. Probiotics & prebiotics: scientific aspects. Wymond- 72. Downes J, Mantzourani M, Beighton D, et al. Scardovia wig-
ham, UK: Caister Academic Press, 2005:1–24. gsiae sp. nov., isolated from the human oral cavity and clinical
53. Bik EM, Long CD, Armitage GC, et al. Bacterial diversity in material, and emended descriptions of the genus Scardovia and
the oral cavity of 10 healthy individuals. ISME J 2010;4:962– Scardovia inopinata. Int J Syst Evol Microbiol 2011;61:25–29.
974. 73. Sundaresan D, Cullinan M, Drummond B, Seymour G, Stanton
54. R^oßcas IN, Siqueira JF. Characterization of microbiota of root J, Heng N. Identification of friend and foe: metagenomics of
canal-treated teeth with posttreatment disease. J Clin Microbiol the oral cavity in health and disease. NZ Medi J
2012;50:1721–1724. 2012;124:1335.
55. Lazarevic V, Ga€ıa N, Girard M, Francßois P, Schrenzel J. Com- 74. Kamalendran N. Oral microbiota of children undergoing com-
parison of DNA extraction methods in analysis of salivary bac- prehensive treatment for severe early childhood caries. 2015;
terial communities. PLoS ONE 2013;8:e67699. Doctor of Clinical Dentistry thesis, The University of Otago.
56. Nyvad B, Crielaard W, Mira A, Takahashi N, Beighton D. 75. Jiang S, Gao X, Jin L, Lo EC. Salivary microbiome diversity in
Dental caries from a molecular microbiological perspective. caries-free and caries-affected children. Int J Mol Sci 2016;17
Caries Res 2012;47:89–102. (pii):E1978.
57. Johansson I, Witkowska E, Kaveh B, Holgerson PL, Tanner A. 76. Jiang W, Ling Z, Lin X, et al. Pyrosequencing analysis of oral
The microbiome in populations with a low and high prevalence microbiota shifting in various caries states in childhood. Microb
of caries. J Dent Res 2016;95:80–86. Ecol 2014;67:962–969.
58. Sanger F, Nicklen S, Coulson AR. DNA sequencing with chain-
terminating inhibitors. Proc Natl Acad Sci USA 1977;74:5463– Address for correspondence:
5467. Professor William Murray Thomson
59. Fleischmann RD, Adams MD, White O, et al. Whole-genome The University of Otago – Sir John Walsh Research
random sequencing and assembly of Haemophilus influenzae
Rd. Science 1995;269(5223):496–512.
Institute
PO Box 647, Dunedin 9054
60. Padmanabhan R, Mishra AK, Raoult D, Fournier P-E. Geno-
mics and metagenomics in medical microbiology. J Microbiol New Zealand
Methods 2013;95:415–424. Email: murray.thomson@otago.ac.nz

24 © 2017 Australian Dental Association

You might also like