Sie sind auf Seite 1von 16

J Clin Periodontol 2017; 44 (Suppl.18): S23S38 doi: 10.1111/jcpe.

12671

Role of microbial communities in Alex Mira1, A. Simon-Soro1 and M. A.


Curtis2
1
Department of Health and Genomics, Center

the pathogenesis of periodontal for Advanced Research in Public Health,


FISABIO Foundation, Valencia, Spain;
2
Institute of Dentistry, Barts and The London

diseases and caries School of Medicine and Dentistry, Queen


Mary University of London, London, UK

Mira A, Simon-Soro A, Curtis MA. Role of microbial communities in the


pathogenesis of periodontal diseases and caries. J Clin Periodontol 2017;
44 (Suppl. 18): S23S38. doi:10.1111/jcpe.12671.

Abstract
: The microbiological characteristics of both caries and periodontal disease show
significant change from those in health. In both instances, there is evidence of co-
association of different organisms into consortia.
Aim: We review and summarize a number of issues pertinent to the community
organization and functional activity of the bacterial populations resident on
supra- and subgingival tooth surface and the influence of these populations on
disease.
Methods: A literature review was undertaken with a particular emphasis on
recent publications involving high-throughput, deep sequencing approaches to the
analysis of microbial populations and their functional activity.
Results: There is increasing evidence to suggest that both caries and periodontal
disease represent dysbiotic states of the oral microbiome. The mode of acquisition
of the oral microbial communities may be less passive than previously recognized
but once established remains relatively stable within an individual although there
are very significant site variations. A repertoire of stable dysbiotic states may
occur in both caries and periodontitis involving different microbial community
structures with potentially similar functional properties. Key words: caries; microbial communities;
Conclusions: The processes which underlie the development and stability of oral microbiome; periodontal disease
microbial populations in the healthy mouth are fundamental to understanding
how these populations are transformed into a dysbiotic state in disease. Accepted for publication 20 December 2016

The biofilms on the supra- and sub- environments. The complexity of flexibility of use to describe either all
gingival surfaces of the teeth are these communities was evident from the species present in a given sample
composed of complex microbial the first time they were subject to or to target specific genera and
communities which have evolved to scientific evaluation some 350 years finally the speed and cost efficiency
inhabit these specialized oral ago by Antonie van Leeuwenhoek of the approach. Its application has
using the very first microscopes and led to the description of 11 phyla in
Conflict of interest and source of
is increasingly appreciated as the the domain Bacteria in the oral
funding statement current methods of high-throughput microbiome in addition to methano-
The authors have stated explicitly and culture independent molecular genic species of the Methanobre-
that there are no conflict of interests methods are applied. Sequence anal- vibacter genus from the domain
in connection with this article. ysis of 16S ribosomal RNA is one of Archaea. The phyla of the domain
Authors Institutions and grant the most recent and most powerful Bacteria that are reliably present
BIO2015-68711-R to AM from Span- of these investigative techniques include Actinobacteria, Bacteroide-
ish MINECO. because of the universal presence of tes, Chloroflexi, Firmicutes, Teneri-
the 16S rRNA gene in all bacteria, cutes, Fusobacteria, Proteobacteria,
2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd S23
S24 Mira et al.

Spirochaetes, Synergistetes and two et al.1994, Lamont & Hajishengallis population level, the inter-individual
currently unnamed phyla, SR1 and 2015) are two well-described exam- variation in microbial community
TM7. Several hundred, distinct spe- ples. More detailed descriptions of structures is also discussed.
cies are contained within these divi- these cooperative and mutually bene- These preceding descriptions aim
sions representing the highly diverse ficial microbial interactions are to provide a contextual background
microbial communities of the mouth described in the paper by Zaura and against which to rationalize the well-
(Human Microbiome Project Con- Marsh (2017, this issue). known shifts in microbial population
sortium 2012). The periodontal Against this background of a structure which occur co-incident
microbiota is particularly heteroge- highly evolved, highly complex and with the alteration from health to
neous, and over 400 species have highly cooperative microbial society, disease in both caries and periodon-
been described in this habitat alone it is pertinent to ask what is the role tal disease as well as the potential
using a 16S rRNA amplification, played by microbial communities in importance of these shifts for diag-
cloning and Sanger sequencing the pathogenesis of the two most nostic and potentially therapeutic
approach (Dewhirst et al. 2010). prevalent diseases at these sites in purposes. Finally, this work ques-
When clonal variation within a bac- the mouth: dental caries and peri- tions whether the changes in micro-
terial species is also taken into odontal disease? It is worth noting bial population structure that are so
account, the composition of these at this point that the overwhelming characteristic of both diseases are
bacterial communities attains an majority of studies of the pathogene- actually responsible for disease initi-
even higher degree of complexity. In sis of these conditions using in vitro ation and progression as opposed to
addition, the fungal oral communi- and animal models and the interpre- inevitable outcomes of the altered
ties have recently been shown to be tation of human studies of the environments that clearly pertain to
very diverse and go well beyond the microbiology in health versus disease a carious lesion or an inflamed peri-
classically reported presence of Can- have taken a reductionist approach: odontal pocket. Is dysbiosis of the
dida species (Ghannoum et al. 2010). the pathological entity is a single microbial communities of the mouth
Fungi have a close interplay with organism or group of organisms act- simply a consequence rather than a
bacteria, and some authors consider ing independently rather than a cause of these diseases?
this interplay beneficial in the main- coordinated, inter-dependent consor-
tenance of oral homoeostasis (Krom tium.
Characteristics of the Microbiology
et al. 2014). However, this manu- In this study, we review and sum-
of Caries and Periodontal Disease
script focuses on the role of bacterial marize a number of issues pertinent
communities, for which information to the community organization and
The microbiology of caries
is more complete. functional activity of the bacterial
The evolutionary adaptations populations resident on the supra- Perhaps influenced by the specific
developed by individual microbes and subgingival tooth surface and aetiology of most infectious diseases,
within these highly diverse communi- the influence of these populations on the characterization of dental caries
ties on the supra- and subgingival disease. In the first instance, a causing agents was reduced for dec-
tooth surfaces will include: the abil- description of the microbiology of ades to mutans streptococci, espe-
ity to adhere; to gain nutrients from both diseases is presented with cially Streptococcus mutans and
the environment for catabolic and particular focus on more recent S. sobrinus (the latter first described
anabolic needs; to evade elimination non-cultivation high-throughput by Louis Pasteur). The epidemiologi-
by microbe and host mediated offen- approaches. The process of acquisi- cal evidence for their association
sive strategies; and to disperse to tion of the oral microbiome is exam- with the disease, its acidogenicity
new environments within and ined as well as the temporal and acidurity is extensive (Loesche
between individuals. However, given appearance and potential source(s) 1986), and in fact, most preventive
that the overriding characteristic of of some of the community compo- strategies against the disease, includ-
life in these oral habitats is in the nents that are frequently associated ing immunization approaches, were
context of a community organiza- with the two diseased states. Recent aimed at these bacteria (Zhang
tional structure, it is likely that many data which challenge the notion that 2014). However, this mutans-centric
of these essential parameters of sur- acquisition is an entirely passive paradigm was challenged when other
vival will be met, either in whole or event are also presented. Next, the acidogenic bacterial species were iso-
in part through a web of community stability of these established bacterial lated from carious lesions and found
interactions. The cooperative degra- communities in health is described to be strongly associated with the
dation (and subsequent consump- with particular reference to the com- disease. These included Bifidobac-
tion) of complex macromolecules by parative resilience of microbial popu- terium (Mantzourani et al. 2009),
the concerted and sequential activity lations at other sites of the human Lactobacillus (Badet & Thebaud
of enzymes produced by different body. Given that a characteristic of 2008) and Scardovia wiggsiae, the
microorganisms and the potential both diseases is their site specificity, latter associated with early child-
protection from host derived antimi- the site variation of the bacterial hood caries (Tanner et al. 2011). In
crobials such as host defence pep- communities both supra- and sub- addition, a sensitive test like PCR
tides and complement by the gingivally within a single individual amplification failed to detect S. mu-
extracellular proteolytic enzyme is also reviewed. To understand the tans in a significant proportion of
activity of a neighbouring member maintenance of health and the cavities (Aas et al. 2008). However,
of the consortium (Bradshaw pathogenesis of disease at the PCR amplification of DNA from
2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Microbial communities S25

carious lesions confirmed the pres- diseases and disease processes which a concrete core of caries causing
ence of many other bacterial species, have a different clinical terminology, agents. This disagrees with the speci-
like Atopobium, Prevotella or but for which specific microbiologi- fic-plaque hypothesis of dental caries
Corynebacterium, among others (Aas cal features must be described. Dif- (Emilson & Krasse 1985) and sup-
et al. 2008, Gross et al. 2012, Tor- ferent carious sites, for instance, ports a view of a non-specific com-
lakovic et al. 2012, Simon-Soro et al. probably imply different microbial munity associated with the disease
2013). Metagenomic studies in which niches, and the same can be true for (Theilade 1986) where an ecological
DNA from carious lesions was cases of chronic and aggressive peri- change triggered by external factors
directly sequenced without any clon- odontitis, or those exacerbated by like sugar promotes the dysbiosis
ing or PCR step also revealed a systemic causes. For example, in (Marsh 2003, Takahashi & Nyvad
complex community, including dif- root caries, where enamel is absent, 2011). Nevertheless, when bacterial
ferent species of non-mutans strepto- a different bacterial composition has composition profiles in different den-
cocci as well as bacteria like been described, with Actinomyces vis- tin cavities were compared, Obata
Veillonella or Capnocytophaga, which cosus or Propionibacterium acidifa- et al. (2014) detected different clus-
appeared to be at high proportions ciens being some of the ters of bacteria. One was domi-
in some cases (Belda-Ferre et al. microorganisms proposed to be asso- nated by Lactobacillus spp., whereas
2012, Sim on-Soro et al. 2013). Thus, ciated with its aetiology (Nyvad & when lactobacilli were absent, a con-
a new picture emerged in which Kilian 1990, Hashimoto et al. 2011, sortia of Propionibacterium, Atopo-
complex bacterial communities Chen et al.2015). Furthermore, when bium and Prevotella could be
appeared to be associated with the different caries lesions are sampled, identified in several lesions. Thus,
disease. The estimated number of dramatic differences in bacterial the co-occurrence of different bacte-
bacterial species in supragingival composition are found, both by rial species that may form part of
dental plaque on teeth sound sur- DNA- and RNA-based methods stable consortia cannot be discarded
faces reached values of 500600, (Gross et al. 2012, Jiang et al. 2013, and more work should be directed in
whereas in dentin cavities decreased Simon-Soro et al. 2014), indicating the future to identify and character-
to 200 and this diversity dropped on that caries aetiology is not only ize those potential cariogenic consor-
average to 125 in non-cavitated polymicrobial but also extremely tia.
enamel white-spot lesions (Sim on- variable among individuals. In addi- Given the strong evidence linking
Soro et al. 2013). Thus, although tion, when different caries lesions mutans streptococci to caries, it was
bacterial diversity was considerably have been sampled from the same surprising to find that S. mutans
reduced under disease conditions, individual, bacterial composition has accounted for <1% of the total car-
probably due to the highly acidic also been shown to vary significantly ies lesion community in both DNA-
environment, the number of bacteria (Simon-Soro et al. 2014). This highly (Gross et al. 2012) and RNA-based
found in caries lesions was consis- variable polymicrobial feature of studies (Sim on-Soro et al. 2014).
tently large, suggesting a polymicro- oral diseases makes the development However, microbial ecology studies
bial aetiology. of vaccines against oral diseases highlight that minorities should not
Given that DNA-based molecular extremely difficult, as a single immu- be underestimated, as bacteria found
methods like PCR can potentially nization target cannot be identified at small proportions could have a
detect inactive or even dead microor- (Simon-Soro & Mira 2015). vital role in the community. In peri-
ganisms, the application of RNA- Nevertheless, some patterns are odontitis, for instance, the presence
based techniques was crucial to con- starting to emerge, and several bac- of P. gingivalis at <1% of the total
firm the presence of complex micro- teria appear to be associated with has been shown to be sufficient to
bial communities in caries lesions. specific lesions or stages. Lacto- develop the disease, by eliciting an
RNA is highly unstable and has a bacilli, for instance, were initially immune response in the host that
short half life and therefore can only associated with advanced stages of allows the establishment of dysbiotic
be detected if bacteria are alive and the disease (Shah et al. 2011), a fea- community of the normally benign
actively growing (Amann et al. ture that has been confirmed by commensal community which ulti-
1995). RNA-based PCR studies iden- molecular methods, which failed to mately are responsible for the intense
tified again dozens of bacterial spe- detect lactobacilli in non-cavitated inflammation (Darveau et al. 2012).
cies on individual enamel or dentin enamel lesions and found this genus Thus, although gum disease is the
caries lesions (Sim on-Soro et al. to be associated with dentin cavities outcome of a microbial dysbiosis,
2014, Sim on-Soro & Mira 2015), (Simon-Soron et al. 2013, Jiang P. gingivalis has been proposed to be
confirming the activity of many bac- et al. 2014). When present, lacto- a keystone pathogen whose activ-
teria that could have a joined or even bacilli appear to be extremely domi- ity is necessary for the development
synergistic effect. When total RNA nant in the lesion, perhaps be of the disease (Hajishengallis et al.
was sequenced by a metatranscrip- displacing other species (Obata et al. 2012). The role of S. mutans as a
tomics approach, the gene expression 2014). In some cases, a few species keystone caries pathogen is more dif-
profiles of individual bacteria could of Lactobacillus accounted for 99% ficult to imagine given the inert nat-
be identified, revealing a complex of the lesion activity (Simon-Soro & ure of the tooth surface, the only
pattern of bacterial activity (Peterson Mira 2015). Except for these cases, region of the oral cavity without a
et al. 2014, Do et al. 2015). different caries lesions appear to har- constitutive mucosal immune
The concept of within-individual bour different bacterial consortia, response. Nevertheless, its potential
variability extends to the various making it extremely hard to identify role in the initiation of ecological
2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
S26 Mira et al.

change by promoting acidification or same genera represented only together in the same sample, has
adherence and triggering the dysbi- approximately 10% of the organisms been interpreted as the existence of
otic state cannot be discarded. From at subgingival diseased sites. Con- consortia of bacterial species acting
an applied viewpoint, the dramatic versely, members of the Bacteroides in concert with one another in mutu-
variation in caries-associated consor- and Fusobacteria were present to alistically beneficial manner. The
tia suggests that traditional antimi- approximately 20% of the subgingi- highly influential study of Socransky
crobial strategies may not be val microbiota but to only approxi- et al. (1998) analysed approximately
effective and new approaches direc- mately 5% supragingivally. 13,000 plaque samples from 185 sub-
ted towards restoration of the eco- This early work has been con- jects using whole genomic DNA
logical balance of dental plaque have firmed and re-emphasized by more probes to 40 bacterial species. Asso-
been proposed (Fejerskov 2004, recent high-throughput non-cultiva- ciations were sought among species
Marsh et al. 2015). tion approaches (see e.g. Colombo using cluster analysis and commu-
et al. 2009). A recent systematic nity ordination techniques. One of
The microbiology of periodontal disease
review of these studies (Perez-Cha- the key and fundamentally impor-
parro et al. 2014) concluded those tant findings of this study, which has
Our understanding of the defining organisms previously shown to be shaped our understanding of peri-
features of the microbiology of peri- periodontitis-associated Porphy- odontal infections ever since, was the
odontal disease has similarly romonas gingivalis, Treponema denti- description of bacterial complexes,
evolved. Nonetheless, there are some cola, Tannerella forsythia, as opposed to individual bacterial
important principles which appear Fusobacterium nucleatum, F. peri- species, that were associated with
irrefutable. These principles can be odonticum, Prevotella intermedia, either periodontal health or peri-
summarized as the 3Cs of the micro- P. nigrescens, Parvimonas micra, odontal disease. The complex most
biology of periodontal disease Com- Campylobacter gracilis, C. rectus, strongly associated with periodontal
munity change, microbial Complexes C. showae, Eubacterium nodatum, disease, the red complex, was com-
and Commensal involvement. Streptococcus constellatus and Aggre- posed of three bacterial species
First, the qualitative and quanti- gatibacter actinomycetemcomitans which subsequently became the focus
tative composition of the microbial (Proceedings of the World Work- of intense investigation: Porphy-
populations in periodontal disease shop 1996, Socransky et al. 1998, romonas gingivalis, Treponema denti-
represents community-wide changes Teles et al. 2013) represented a far cola and Tannerella forsythia. Other
to the composition in health. These too restricted list. On review of 41 complexes, for example the yellow
changes represent global shifts in the different studies, they provided evi- complex which comprised predomi-
overall population structure and dence to support the potential nantly different streptococcal species,
include the emergence of organisms involvement of 17 new additional and the green complex which con-
not normally encountered in health candidate organisms based on their tained a preponderance of capnocy-
and a disappearance or reduction in reported association with periodonti- tophaga species, represented early
others. The cultivation studies of tis in 35 independent investigations. colonizers of dental plaque which
Moore and colleagues some thirty These organisms include several were more closely associated with
years ago (Moore et al. 1982) pro- members of bacterial phyla previ- health. The orange complex con-
vide a convincing demonstration of ously implicated (Bacteroidetes, Fir- tained those organisms generally
this principle. In this investigation of micutes, Proteobacteria, Spirochaetes considered to colonize dental plaque
the bacteriology of severe general- and Synergistetes), a new phylum later: fusobacteria species, members
ized periodontitis in 21 individuals, (Candidatus Saccharibacteria) and of the prevotella and the campy-
they described the isolation and also members of the Archaea lobacter. More recent deep sequenc-
characterization by biochemical tech- domain of life, which has not previ- ing approaches have expanded these
niques of 2723 individual isolates ously been associated with any infec- original observations (e.g. Griffen
representing 190 bacterial species, tious disease of humans. This et al. 2012). In a recent study, Kirst
subspecies or serotypes. Of these, 11 seemingly endlessly increasing list of et al. (2015) used 16S rRNA
species exceeded 1% of the subgingi- disease-associated organisms adds sequencing to survey the subgingival
val flora and were most closely asso- further weight to the notion that the microbiota in 25 individuals with
ciated with disease and 11 others changes in the microbiology of peri- periodontal disease and 25 healthy
were also sufficiently frequently iso- odontal diseases represent a commu- controls. Through cluster analysis
lated to be deemed suspected agents nity-wide perturbation of the entire techniques, they demonstrated the
of tissue destruction. This study microbial population structure and presence of two predominant clus-
highlights the marked difference function of subgingival plaque (Cur- ters. One cluster characterized by
between the overall microbiota in tis 2014). high levels of Fusobacterium and
periodontally diseased, subgingival The second principle which has Porphyromonas bacterial species was
sites compared to the adjacent emerged from the study of microbial strongly associated with the diseased
supragingival microbiota. Whilst the populations in periodontal disease is population, whereas a second clus-
supragingival microbiota was domi- the clear co-association of different ter, this time dominated by Rothia
nated by the Actinomyces spp., organisms in the microbiota in and streptococci was representative
Streptococcus spp and Veillonella health and disease. This co-associa- of periodontal health. Analogous to
spp., which comprised some 40% of tion, the tendency of some groups of different gut enterotypes, based on
the total cultivatable bacteria, the organisms to be consistently detected the abundance of key bacteria in the
2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Microbial communities S27

gut microbiota, the authors sug- Acquisition of Oral Microbial


has been shown to influence the sub-
gested that these discrete clusters in Communities
sequent development of a healthy
periodontal health and disease may gut microbiota, where colonization
represent different periodonto-types Some understanding of the temporal by some key components of the
which differentiate between peri- acquisition of microbial populations microbiota, such as the Bacteroides,
odontal health and disease. Hence, and the development of the complex is significantly delayed in babies
as might be predicted from the meta- oral biofilms in the mouth (Gibbons born by C-section (Rutayisire et al.
bolic and cooperative interactions 1989 K on
onen et al. 1999) may be 2016), and a similar process may
described in the paper by Zaura and required in order to determine the occur in the oral cavity. For
Marsh (2017), discrete consortia of susceptibility of these populations to instance, using a DNA chip, three-
bacterial species within the overall dysbiosis. These steps in the develop- month-old babies born by C-section
complex microbial population struc- ment of the oral microbiome to a were found to harbour a reduced
ture appear to be the norm in dental mature a stable health state are illus- bacterial diversity in the mouth (54
plaque in health and disease. trated in Fig. 1. species) compared to vaginally deliv-
A third emerging principle con- The conventional view of bacte- ered children (79 species), with some
cerns the contributory role of organ- rial transmission to the child bacteria like Slackia exigua being
isms normally considered to be involves initial passive transfer exclusively found in the latter (Hol-
commensal or health associated. The from the mother through the birth gerson et al. 2011). Very recently, it
absolute requirement of the com- canal. This view has been confirmed has been shown that experimental
mensal microbiome in order for by high-throughput sequencing data oral exposure of babies born by C-
P. gingivalis to induce periodontal in children born by vaginal and cae- section to maternal vaginal fluids
disease in the murine model sarean delivery (Dominguez-Bello increases the colonization of vaginal
(Hajishengallis et al. 2011) under- et al. 2010). The microbial composi- bacteria in the oral cavity, the gut
scores the functional importance of tion of the gut of babies born via and the skin of the babies, and par-
the microbial community associated C-section resembled the mothers tially restores their oral microbiota,
with health to the progress of dis- skin bacteria whereas babies born which shifted towards that found in
ease. Furthermore, the results of via conventional delivery had differ- babies born by vaginal delivery
transcriptional profiling of microbial ent communities with some similarity (Dominguez-Bello et al. 2016). Given
samples from periodontally healthy to the vaginal microbiota. In longi- the higher epidemiological risk of
versus diseased sites (Duran-Pinedo tudinal studies of faecal samples, the immune-related diseases in children
et al. 2014b) have demonstrated that initial bacterial composition at birth born by C-section (Huurre et al.
the majority of potential virulence
factors upregulated in human disease
are derived not from the traditional
group of periodontal bacteria but
instead from those bacteria normally
associated with health. This may be
explained by the effect of altered
environmental conditions or, poten-
tially, by a direct effect of a peri-
odontal bacterium such as
P. gingivalis on the pattern of gene
expression of neighbouring organ-
isms as demonstrated in mixed bio-
film in vitro studies (Frias-Lopez &
Duran-Pinedo 2012, Duran-Pinedo
et al. 2014a). Hence, our classifica-
tion of an organism as a commensal,
and therefore invariably in harmo-
nious balance with the host, is
almost certainly too rigid a defini-
tion. The key determinant to
describe the association of any bac- Fig. 1. Compositional transitions in the oral microbiome. The microbial communities
terium with health (or disease) is dic- of the mouth undergo compositional changes from birth and throughout development.
tated by the functional activities Health is associated with a relatively stable community structure which is largely resili-
produced by that cell at any given ent to change by external stresses. Dysbiosis occurs when a threshold in the resilience
mechanisms is overcome due to dietary influences in caries and predominantly,
time and these may change depen- although not exclusively, host factors in periodontal disease. The dysbiotic state dis-
dent upon the environmental context plays functional differences which contribute to disease and may also be resistant to
including the presence of other bac- compositional change. Acute changes may also occur in health leading to an unstable
teria with the ability to manipulate semi-dysbiotic state which can be reversed to the original healthy community structure
the transcriptional profile of commu- or alternatively undergo further compositional change to a stable dysbiosis. Adapted
nity neighbours (Darveau et al. from Lozupone CA, Stombaugh J, Gordon J, Jansson JK, Knight R. Nature. 2012
2012). 489(7415):220230.

2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
S28 Mira et al.

2008), together with the important delivery mode and perinatal factors developing foetus may be exposed to
role played by the immune system (Li et al. 2005), and have proposed a microorganisms before birth. This
in oral diseases (Silva et al. 2015), link with the onset and severity of new concept is based on the identifi-
it is not unreasonable to think that caries disease (K ohler et al. 1988, Li cation of a microbiota in the pla-
acquisition of an aberrant micro- et al. 1994, Lai et al. 1997). In other centa which appears to resemble that
bial composition at birth may have studies, the oral microbiota at of the oral cavity (Aagaard et al.
longer term consequences on the 3 months of age could not be related 2014) and to be influenced by pre-
development of dysbiotic oral to caries development at 3 years term delivery and by infections dur-
microbial communities. (Holgerson et al. 2015). Finally, not ing the first term of pregnancy
After this initial colonization pro- only disease-associated species but (Prince et al. 2016). In a study of
cess, sequential development has tra- also health-associated ones have been infants born by C-section, the bacte-
ditionally been shown to be governed detected in longitudinal studies, like rial composition of the placenta
by saliva exchange and other poten- Neisseria flavescens and Porphy- appeared to resemble that of the
tial environmental sources. This is romonas catoniae, which have been amniotic fluid and the meconium of
supported by work showing related- proposed as biomarkers of caries-free the infant (Collado et al. 2016)
ness of infant bacterial strains to children (Crielaard et al. 2011). The opening up the possibility that the
family members or children care- acquisition of oral anaerobes has infants were exposed to a given set
givers from whom microorganisms been addressed by K ononen et al. or microorganisms in utero.
may have been transmitted (Li & 1999, and suggests again the colo- Although no oral samples were
Caufield 1995), although this associa- nization of given species in a sequen- taken in this experiment, faecal
tion has been shown to be weaker tial, timely manner. material from the infant at 3 days of
than initially expected (Cephas et al. In recent years, driven by the age had a similar bacterial composi-
2011). In addition, the dramatic application of high-throughput tion to that of breastmilk, suggesting
changes in the anatomy of oral tis- sequence analyses, two important that the oral cavity could also
sues throughout childhood, the new concepts have emerged in addi- undergo a similar process of micro-
immune response and dietary tion to these established views of bial succession to that reported in
changes have long been known to passive acquisition of oral microbial the gut. It has to be kept in mind,
influence microbial composition. For communities. First, it is proposed however, that the microbial load
instance, the eruption of teeth has that human breastmilk harbours sev- detected in these samples is extre-
been proposed to influence the colo- eral hundred bacterial species that mely low and therefore the possibil-
nization by microorganisms, espe- change from colostrum through ity for contamination at the moment
cially those living on hard tissues, mature milk. This microbiota of sampling, sample processing and
and an increase in bacterial diversity includes many oral microorganisms DNA extraction cannot be excluded,
has been observed at this point such as streptococci, Veillonella or as even the traces of DNA in com-
(Rotimi & Duerden 1981, Brailsford Leptotrichia (Cabrera-Rubio et al. mercial kits have been shown to be
et al. 2005). To these environmental 2012, Jeurink et al. 2013). The sufficient to provide positive bacte-
parameters, we have to add the breastmilk-associated microbiota has rial PCR amplification (Biesbroek
potential impact of prosthodontic/ been shown to be transmitted to the et al. 2012, Lauder et al. 2016).
orthodontic appliances, which have a infant gut (Collado et al. 2016), and Thus, the potential presence of
profound impact in bacterial colo- therefore, the same would be microbes in utero would appear to
nization (Koopman et al. 2015). In expected to happen in the oral cav- be at really low densities that could
the case of well-studied oral patho- ity. In fact, although oral bacteria be confounded with contamination,
gens like mutans streptococci, a could be transmitted from the and appropriate quality controls, as
window of infectivity was proposed infants oral cavity to the mammary well as careful sample handling
around the time of first teeth erup- glands during breastfeeding, the should be performed to establish
tion for initial colonization (Caufield appearance of oral bacteria in pre- whether there really is a pre-birth
et al. 1993), and also for subsequent colostrum suggests that breastmilk microbial exposure.
periods of teeth eruption, like those could in fact be an important source If the presence of microorganisms
of primary molars or permanent first for directed microbial colonization in utero is confirmed, the conse-
molars (Brailsford et al. 2005). How- of the oral cavity (Mira & Rodriguez quences for the development of the
ever, several manuscripts have chal- 2016). Given that different oral infant immune system and for oral
lenged this concept, as mutans microbial profiles have been found diseases will also be important. A
streptococci have been found in between breastfed and formula-fed change of paradigm has been pro-
infants before teeth eruption (Mil- infants and that some bacteria asso- posed in which the placental tro-
grom et al. 2000, Wan et al. 2001). ciated with breastfeeding have been phoblasts (main constituents of the
In addition, other authors have found to inhibit the growth of oral placenta), which have been shown to
found a constant increase in mutans pathogens (Holgerson et al. 2013), respond to bacterial components
streptococci with time without a dis- the influence of breastfeeding on the (Koga et al. 2009), would interact
crete window of infectivity (see Law risk of caries and periodontitis is an with commensal microorganisms,
et al. 2007 for a review). This could area for further investigation. which could induce tolerance rather
be relevant for disease, because some Secondly, although the topic is than rejection (Mor & Kwon 2015),
authors have found a correlation of not without controversy, several especially if their antigens are pre-
S. mutans initial colonization with authors have proposed that the sent in small amounts (Sabatos-
2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Microbial communities S29

Peyton et al. 2010). Thus, the poten- specific to each of the multiple regarding intra-individual variability
tial presence of antigens from oral niches, for example, the tongue, in fungal composition.
bacteria in the placenta has been cheek mucosa, dental tissues, gingi- It is interesting to note that when
proposed to develop foetal tolerance val crevice or hard palate (Fejerskov saliva has been collected from the
towards the maternal oral micro- et al. 1994, Babaahmady et al. 1997, same individuals for which supra-
biome during pregnancy (Zaura Aas et al. 2005, Zaura et al. 2009). and subgingival dental plaque sam-
et al. 2014). Co-incidentally, many The initial work, performed by cul- ples were available, very different
physiological changes during human turing, DNA hybridization and bacterial profiles were obtained (see,
pregnancy facilitate bacterial translo- molecular cloning of the 16S gene for instance Segata et al. 2012,
cation into the maternal circulation, has been confirmed by the enormous Sim on-Soron et al. 2013). This has
including increased mucosal perme- effort carried out by the Human important applied value, because sal-
ability and gum inflammation (Mira Microbiome Consortium, which iva has traditionally been a preferred
& Rodriguez 2016). Furthermore, sampled several hundred individuals sample for epidemiological and even
injection of oral samples (saliva and at different body parts, including six aetiological studies, due to its non-
plaque) into pregnant mice resulted oral locations and analysed them by invasive nature and its convenience.
in colonization of placenta by oral massive sequencing and metage- This could be the reason why some
microorganisms (Fardini et al. 2010). nomic techniques (Human Micro- studies have found an association
In this way, the immune system of biome Project Consortium 2012). between microbial composition and
the new born infant would be condi- However, within specific tissues, sev- disease in plaque samples but not
tioned to accept the normal micro- eral authors have also unravelled sig- saliva, in both dental caries and gin-
biome of the mother in preference to nificant heterogeneity in microbial givitis (Ling et al., 2010; Huang
bacteria from other sources. This composition. This was not unex- et al., 2011), as saliva contains
interesting hypothesis would have pected because the earlier classical microbes from all oral locations,
important consequences, as the studies had already shown striking including those not related to the
maternal oral health status during changes in environmental features disease. This probably introduces a
pregnancy could directly influence like pH between different teeth large microbial diversity which
the acquisition, post-partum colo- (Kleinberg & Jenkins 1964), and undermines the diagnostic value of
nization and immune recognition of changes in oxygen availability, redox salivary microorganisms and high-
the infants oral microbiome. This potential, chewing forces and even lights the importance of appropriate
could be especially relevant for preg- hygiene differences, among others, sampling for microbial-based diag-
nant women with periodontitis, as have been proposed to influence and nostic tests of caries and periodonti-
the presence of periodontal patho- fragment a given oral site into multi- tis (Belda-Ferre et al. 2015).
gens in breastmilk and placenta ple micro-niches (Zaura et al. 2009). The dramatic level of variability
could be facilitated, increasing infant When different teeth were sampled in microbial composition detected
immune tolerance to a disease-prone at the same mesio-buccal location, between healthy sites, and also
microbiota. Supporting this view, a astonishing differences were between disease sites, within the
recent qPCR-based study has observed in bacterial composition, same individual underscores the
detected several periodontal patho- with twenty species being associated importance of proper lesion classifi-
gens in placenta, which were found with specific tooth locations (Haffa- cation for microbiological studies
at higher levels in samples from jee et al. 2009). In another study, and that an important effort must be
mothers with periodontitis (Blanc Sim on-Soron et al. (2013) sampled put into appropriate sampling at
et al. 2015). From an applied point plaque from over 100 locations per specific sites (discussed by Nyvad
of view, the confirmation of the pla- individual, observing significant dif- et al. 2013), as opposed to the fre-
cental microbiota-mediated tolerance ferences in bacterial composition quent pooling of material for subse-
would have consequences for public between teeth of the same individual quent analysis. This includes also the
health policies directed towards and above all between vestibular and distinction between active and inac-
improving oral health in pregnant lingual surfaces of the same tooth. A tive caries lesions, clinically diag-
and breastfeeding mothers, as this repeated feature included a clear nosed by texture, brightness and
could reduce the prevalence of oral increase of streptococci at the colour, but not fully characterized at
diseases of children later in life. Nev- vestibular surfaces of tooth and gin- the microbiological level (Nyvad &
ertheless, more longitudinal studies gival sulci, where they accounted on Fejerskov 1997).
are required to provide solid epi- average for 48% and 43% of all bac- The microbiology of dental caries
demiological evidence showing that teria, whereas on the lingual sur- and periodontitis at different stages
early use of antibiotics, mode of faces, they reached only 24% and of the disease has shown that they
delivery, breastfeeding habits and 12% in supra- and subgingival pla- are both better understood as a
other factors impacting early life que samples, respectively. In addi- dynamic process, and not as a classi-
microbiota acquisition are linked to tion, Streptococcus and cal infectious disease with a well-
oral diseases later in life. Fusobacterium appeared to have defined aetiology. In dental caries,
opposite frequency patterns, suggest- different microbial communities have
ing adaptation of individual bacteria been found at the initial (enamel-
Within-individual Variability
to specific microniches (Simon-Soron degrading) and advanced (dentin
The oral cavity harbours different et al. 2013) (Fig. 2). Unfortunately, expansion) stages (Gross et al. 2012,
microbial communities which are there is very limited information Sim on-Soro et al. 2012, Sim on-Soro
2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
S30 Mira et al.

study, there appeared to be a bacte-


rial species which was present in all
samples, namely Streptococcus mitis,
in agreement with other DNA-based
studies performed with a lower num-
ber of samples (Aas et al. 2005). In
addition, another 13 genera were
found in more than 50% of the sam-
ples. Utter et al. (2016) also detected
that a few bacterial genera
accounted for most of the dental
plaque community in American and
Chinese populations, and the differ-
ent genera varied in their stability,
which fluctuated around a given
mean. Although bacterial composi-
tion was found to be fairly stable at
Fig. 2. Intra- and inter-individual variability in oral microbiota composition. Bar
the genus and species level through
graphs show the proportion of different bacterial genera in supragingival and subgin-
gival dental plaque from two healthy individuals in the same molar (tooth 4.6), col- time, a nucleotide resolution level
lected either at the vestibular (buccal) or lingual surfaces. DNA was extracted from analysis revealed that the proportion
the samples, which were subject to pyrosequencing of the 16S rRNA gene to deter- of different strains varied within
mine bacterial composition. Important differences are found at the same site and loca- each bacterial taxa, and that some of
tion between the two individuals, as well as between supra- and subgingival plaque these strains could be described as
samples. In addition, large differences in the proportion of some bacteria were found bacterial fingerprints in dental pla-
between the vestibular and lingual surfaces of the same tooth. For instance, Strepto- que, that is individual-specific strains
coccus dominated the vestibular surface of tooth and gingival sulcus in individual 2, fluctuating around a stable mean
but its proportion was significantly lower on the lingual surface, where Fusobacterium over time. For instance, over a
was the most frequent organism in this individual. The data illustrate the enormous
variability in microbial populations not only between individuals but also within well-
dozen Corynebacterium strains were
defined intra-oral niches. Sequences were downloaded from Sim on-Soro et al. 2012. detectable in each individual, of
which a different subset reached high
abundance in any given person. In
another study performed in adults,
& Mira 2015, Obata et al. 2014). In with early childhood caries lesions significant differences were found in
some cases, both carious lesions at (Tanner et al. 2011) but had passed the estimated salivary bacterial den-
different stages and samples from previously unnoticed in adult sity over a period of 1 year, suggest-
sound surfaces of the same individ- cavities. In adolescent aggressive ing possible seasonal changes
ual were available and confirm a dif- periodontitis, Aggregatibacter actino- (Cameron et al. 2015). However,
ferent bacterial composition, mycetemcomitans has been identified salivary microbiota composition in
indicating that the shift in micro- 
as a causative agent (Aberg et al. those individuals was also found to
biota is not due to inter-individual 2015) and other bacteria potentially be fairly stable. These features con-
differences (Sim on-Soro et al. 2014). involved in this disease are being trast to other human body habitats
The same was demonstrated in an examined (Clerehugh & Tugnait like the gut, where a higher degree
induced gingivitis cohort where 2001, Oh et al. 2002, Shaddox et al. of temporal variability was found
all 20 patients in the study developed 2012, K on
onen & M uller 2014) and and a where clear core, conserved
the disease after 14 days of compared to the classical disease composition could not be found
absence in toothbrushing (Kistler complexes in adult forms of the (Turnbaugh et al. 2009). This is sur-
et al. 2013). In these individuals, disease. prising given the high exposure of
subgingival bacterial composition at the oral cavity to external contami-
baseline (healthy gingival sulcus) and nation and shows that the oral
Stability of the Microbial
gingivitis within the same patients microbiome has an important degree
Communities in Health
was studied by massive sequencing of stability under health conditions.
of 16S rRNA genes, detecting differ- Stahringer et al. (2012) analysed the Certain stability in the oral
ences in composition and activity DNA sequences of saliva samples microbiota has also been observed in
through time. Bacterial taxa associ- (representing bacteria from different shorter time periods when compared
ated with gingivitis included mouth locations) from over 200 ado- to other human niches. For instance,
Fusobacterium nucleatum subsp. poly- lescents over a 10-year period and tongue samples from two individuals
morphum, Lachnospiraceae sp., Lau- detected a core salivary microbiota were collected on a daily basis for a
tropia sp. and Prevotella oulorum. that can be defined by eight bacterial one-year period (Caporaso et al.
Variability of caries and peri- genera which were detected in >95% 2011). Bacterial diversity was deter-
odontitis microbial aetiology at dif- of all samples. These included Strep- mined by pyrosequencing, and its
ferent ages must also be better tococcus, Veillonella, Gemella, Gran- analysis showed that the tongues
studied, as specific organisms like ulicatella, Neisseria, Prevotella, microbiota was clearly distinguish-
S. wiggsiae have been associated Rothia and Fusobacterium. In this able from that of other body parts
2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Microbial communities S31

from the same individual, such as time, which showed similar degrees driven pathologies and this can be
the gut or the skin. The fact that of intra-individual variation in extended to caries and periodontal
oral samples normally appear microbial composition in monozy- disease (Fig. 1). Whilst no single
together in principal components gotic and dizygotic twin pairs (Stah- organism or collection of organisms
analyses (PCAs) when other human ringer et al. 2012). In fact, twins have been identified as a consistent
samples are included suggests that salivary bacterial composition was marker in any human-associated
oral microbial communities are more more similar to each other than to microbiota, several defining features
stable than other body habitats. the whole population at all time- of dysbiosis have emerged through
However, this does not imply that points, but become more different investigation of these pathology-
there is low intra-oral variability. In when they aged and no longer associated microbial populations in
fact, a large variation was observed cohabited. Lifestyle was also found the last decade.
within these tongue samples through to be the driver of variability in First, in several examples, dysbio-
time, as only 10% of the species other studies (David et al. 2014). sis is reflected in a reduction in the
were estimated to be part of the Thus, the data suggest that it is the overall microbial diversity of the cor-
stable or core microbiome. Expo- environment, and not only the responding symbiotic community.
sure to different foods, medication, genetic makeup, which accounts for For example, a reduction in taxo-
travel and changes in immune the large inter-individual variability nomic diversity and species member-
response, among others, were and for perturbations in within-indi- ship of the microbiota has been
hypothesized to be linked to changes vidual stability. observed in multiple studies of the
in microbial composition, including human gut microbiota in disease in
blooms of individual taxa that were addition to animal infection models
The Concept of Symbiosis versus
frequently observed (Costello et al. (Pham & Lawley 2014). Dysbiosis in
Dysbiosis
2009). Thus, from a practical view- the gut generally leads to a depletion
point, defining an individuals micro- The overall conclusions of the stud- of obligate anaerobic bacteria such
biota based on the results obtained ies listed above are that the composi- as Bacteroides and Ruminococcus
from a single sample may not be tion and structure of oral microbial spp., and conversely, an increase in
representative and will be influenced communities are flexible throughout facultative anaerobes including the
by the relative presence of transient development but attain a relatively family Enterobacteriaceae (e.g.
community members. stable state more stable than gut, Eschericia coli, Klebsiella spp., Pro-
Although more longitudinal stud- the nares and the skin (Costello teus spp.). Manichanh et al.(2006)
ies are needed to address the ques- et al. 2009). Equally, they are reported a decrease in microbial
tion of oral microbial composition responsive to the lifestyle of the indi- diversity in the faecal microbiota in
stability throughout life, current data vidual and to environmental effects Crohns disease. Lepage et al.(2011)
suggest a progressive increase in which can elicit changes in composi- reported a similar reduction in ulcer-
complexity and diversity from birth tion and abundance of different spe- ative colitis, and Chang et al.(2008)
to the first years of life, with impor- cies over both extremely short and described decreased diversity of the
tant alterations during teeth erup- prolonged timescales. Shifts or dif- faecal microbiota in recurrent
tion, end of breastfeeding, ferences in the population structure Clostridium difficile-associated diar-
introduction of solid food, perma- of human-associated microbiota do rhoea. A functional consequence of
nent teeth eruption and maturation not necessarily equate to a dysbiosis, a less diverse gut microbiota appears
of the immune system. Oral micro- rather they are representative of the to be a reduced metabolic capacity,
biota during adolescence and adult- flexibility of these populations which exemplified by a decline in short
hood appears to be more stable than are entirely compatible with health. chain fatty acid production. These
in other well-studied niches like the On a more general note, given the physiological by-products of carbo-
gut, but still with an important apparent flexibility of the microbiota hydrate fermentation by the micro-
degree of within-individual variabil- associated with humans, there must biota are important energy sources
ity. That variability occurs in the be equal levels of flexibility in the for the host and also enhance the
first place due to dental plaque recognition and response systems mucosal barrier, inhibit intestinal
development from the moment of which interface with these micro- inflammation and oxidative stress.
toothbrushing, as the dental biofilm biota, the full details of which have Hence, a reduction in the metabolic
increases in complexity within hours yet to be elucidated. capacity of the microbiota may con-
to a mature state in a few days If flexibility represents the norm, tribute to the impairment of host
(Kolenbrander et al. 2006). But there how is dysbiosis defined? The critical defences and thereby promote the
are also important intra-individual differentiating factor is the response stability of a dysbiotic community
changes in microbiota composition of the host. Dysbiosis is now recog- (Pham & Lawley 2014).
at a longer scale, which suggests that nized as a definitive change in the The microbial changes that are
environmental effects are the drivers microbiota at a given site in the characteristic of the development of
of that variability. Whether the envi- body, crucially, accompanied by a dental caries adhere to this feature
ronment or human genetic back- breakdown of hostmicrobial mutu- of dysbiosis: the overall richness of
ground is behind the large inter- alism. Dysbiosis of human-asso- the supragingival community is
individual variability was elegantly ciated microbiota is now thought to diminished as those bacteria with a
addressed by studying saliva samples be the defining event of multiple more acidophilic nature predominate
in a set of 45 twin pairs through inflammatory and systemically whilst those less able to tolerate low
2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
S32 Mira et al.

pH are reduced. However, the descending from the cemento-enamel is not to diminish the potentially
obverse appears the case in peri- junction to the migrating front of pivotal role played by individual
odontal disease. Here, the overall the pocket epithelium. Perhaps this organisms in driving dysbiosis in a
diversity of the subgingival microbial diversity of niches is then reflected in host-associated microbiota. Recent
population frequently is increased the increased richness of the micro- observations using the mouse model
relative to that in health (Griffen et bial populations. of periodontal disease have demon-
al. 2012, Camelo-Castillo et al. The second emerging feature of strated that one of the presumed
2014), although this view has been dysbiotic communities is a preferen- important pathobionts of the dis-
challenged in other investigations tial loss of organisms considered ease, Porphyromonas gingivalis may
(Kirst et al.2015). The apparent beneficial to human health and a contribute to disease by altering the
divergence from the norm in the case corresponding increase in patho- normal oral microbiota to a dysbi-
of periodontal disease may have bionts, members of the normal com- otic state (Hajishengallis et al.2011).
underlying significance for the mensal microbiota with the potential These studies in mice demonstrated
aetiopathogenesis of the disease. to cause pathology. In the case of that the oral commensal microbiota
What might provide a mechanis- dysbiosis of the gut, these changes is responsible for the tissue and bone
tic explanation for the distinction are exemplified by well documented destruction associated with peri-
between decreased diversity in dys- reductions in the levels of obligate odontitis when just low numbers of
biosis in caries versus increased anaerobic members of the Firmicutes P. gingivalis are present. This organ-
diversity in periodontal diseases? phylum and increased representation ism was accordingly described as a
One possibility is that the environ- of members of the Proteobacteria, in community activist able to orches-
ment of a diseased periodontal particular the family Enterobacteri- trate the normal benign periodontal
pocket may represent a site of aceae (recently reviewed in Walker & microbiota into a dysbiotic commu-
immune dysfunction which permits Lawley (2013) and Hold et al. 2014). nity structure (Darveau et al. 2012).
the proliferation of species normally Most human pathogens belong to
controlled by the host defence. Low- the Proteobacterium phylum includ-
Functional Properties of the
ered efficiency of the innate and ing members of the Enterobacteri-
Microbial Communities in Caries and
adaptive response in periodontal dis- aceae which contains a number of
Periodontal Disease
ease due to host or bacterially medi- frank and opportunistic pathogens
ated effector mechanisms is not including Salmonella spp., Shigella The polymicrobial nature of peri-
inconsistent with many in in vitro spp., Klebsiella spp., Proteus spp. odontitis and dental caries not only
and animal investigations. Inhibition and E. coli. Similarly in caries and implies that several organisms can be
of IL-8 activity by P. gingivalis so- periodontal disease, marked shifts in responsible for the onset of disease,
called chemokine paralysis (Darveau the microbial population structure but also that they could have syner-
et al.1998) provides one such exam- are observed as described in a pre- gistic effects. Synergy in relation to
ple. Support for this mechanistic ceding section. What is less clear in virulence has been demonstrated in
explanation comes from a surprising the case of these two conditions, several complex microbial communi-
source: the mucosal microbiota asso- however, is which organisms or con- ties, ranging from abscesses to otitis
ciated with colorectal cancer sortia of organisms we should con- media or chronic wounds, as well as
tumours and polyps sites of sider to be beneficial organisms and oral diseases (Murray et al. 2014). In
immune suppression in the colon which should be considered the dental caries, an example is given by
also demonstrate elevations in popu- pathobionts. Identification of the consortia between Veillonella alcales-
lation diversity compared to healthy former may have significant thera- cens and S. mutans. Veillonellae and
mucosal surfaces (Mira-Pascual et al. peutic potential for both conditions. streptococci appear together in
2015). An alternative explanation For example, new bacterial species microscopy images of dental plaque
may be simply that the increased with antimicrobial and pH buffering and coaggregate in vitro (Hughes
nutrient supply due to the ingress of capacity have been found to be asso- et al. 1992, Dige et al. 2014), which
gingival crevicular fluid in periodon- ciated with caries-free conditions and has been interpreted as a metabolic
tal disease provides such a rich envi- have been proposed to be used as dependency, because S. mutans pro-
ronment for enhanced bacterial oral health promoting probiotics duces lactate as a result of sugar fer-
growth rate that this is sufficient to (Camelo-Castillo et al. 2014, Huang mentation and Veillonella utilizes
outweigh even the very significant et al. 2016) and a similar strategy lactate as carbon source (Mikx &
challenge presented by the elevated has been followed for periodontal van der Hoeven 1975). The interest-
inflammatory response. However, disease (Teughels et al. 2011). ing feature is that when these species
given that an elevated nutrient sup- The overriding consensus of were grown together in an artificial
ply is also likely to be a feature at investigations of dysbiotic micro- mouth model, they were shown to
sites of disease in inflammatory dis- biota in human disease is that rather produce more acid than any one of
orders of gut, where microbial diver- than seeking to describe a single bac- them separately (Noorda et al.
sity decreases compared to health, terium or even group of organism 1988), suggesting a synergistic cario-
this explanation appears to have less responsible for the disease in a genic effect.
credibility. Finally, one could argue hugely complex microbial system, Similarly, there are a growing
that a diseased site in the periodon- the microbiota as a whole should be number of examples of synergistic
tium actually represents multiple viewed as the pathological determi- interactions between different organ-
geographically dispersed niches nant. Whilst this may be the case, it isms in the periodontal microbiota
2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Microbial communities S33

which may contribute to disease. (Bentez-P


aez et al. 2014). In addi- confirm that these microbial collage-
Animal studies over the last 20 years tion, although the presence of some nases can degrade the important
have consistently demonstrated that bacteria, like Fusobacterium, was protein component of dentin.
poly-microbial infections with, for found to be similar in healthy gingi- In the case of periodontitis, whole
example, combinations of P. gingi- val sulci and periodontal pockets, its RNA and DNA sequencing has been
valis, T.denticola, T. forsythia and pattern of gene expression was found performed on healthy and disease
F. nucleatum cause significantly to be radically different between the sites, which has allowed identifying
higher levels of pathology than two conditions (Yost et al. 2015). those functions overexpressed under
mono-infections with these bacteria This indicates that the microbial each condition (Duran-Pinedo et al.
(Feuille et al. 1996, Ebersole et al. community as a whole behaves in a 2014b, Yost et al. 2015). These stud-
1997, Kesavalu et al. 2007, Verma different way under both conditions, ies show a large diversity of active
et al. 2010a, b). Some mechanistic and emphasizes the importance of functions in the healthy gingival sul-
explanations for these synergies are functional analysis, which are now cus, whereas periodontal pockets
now emerging: for example S. gor- possible due to the advances in showed a congruence of several gene
donii may enhance the virulence of metagenomic (whole DNA analysis, functions that appeared to be over-
Aggregatibacter actinomycetemcomi- providing the genetic repertoire of expressed in all patients. One of the
tans by a cross-feeding mechanism the microorganisms) and metatran- activities which could be influencing
(Stacy et al. 2014); T. denticola and scriptomic (whole RNA analysis, virulence was that of gingipain, a
P. gingivalis have the potential to providing the expression pattern of protein synthetized by P. gingivalis
cooperate metabolically (Tan the community) approaches (Nyvad which cleaves the complement com-
et al.2014); Prevotella intermedia and et al. 2013). ponent 5 (C5) generating high levels
P. gingivalis can act synergistically in In enamel caries lesions, for of C5a locally, which has a potent
the acquisition of haemin from hae- example, DNA-based functional inflammatory effect (Hajishengallis
moglobin (Byrne et al. 2013). studies have shown an over-represen- et al. 2012). Other overexpressed
These in vitro and animal model tation of genes encoding pH stress functions in chronic periodontitis
studies are now being complemented proteins, as well as enzymes for the included biological processes related
by data from human studies on the degradation of dietary sugars, which to flagellar motility, peptide trans-
functional activities of oral microbial is consistent with the established role port, iron acquisition, beta-lactam
consortia in health and disease. Until of cariogenic microbes to develop a degradation and biosynthesis of the
recently, most studies on the oral cavity. However, when dentin caries lipid A component of endotoxins
microbiology of caries and periodon- lesions were studied, over-repre- from Gram-negative bacteria. In one
titis have focused on the bacterial sented genes included those for study, a metatranscriptomic analysis
composition of samples under health osmotic stress, allantoin degradation, of active and inactive sites from
and disease conditions, showing that proteolytic activity, and utilization patients with chronic periodontitis
both diseases involve a clear dysbio- of glycans, among others (Belda- was performed (Yost et al. 2015).
sis, or change in the balanced pro- Ferre et al. 2012, Sim on-Soron et al. The over-represented functional sig-
portion of different microorganisms. 2013). This suggests that the micro- natures at active, progressing sites
However, the composition of micro- bial communities in initial, enamel included cell motility, transport (ci-
bial consortia associated with health lesions perform different functions trate, iron, potassium, chloride and
and disease varies considerably than those of more advanced, dentin amino acids), lipid A and peptido-
between individuals and between cavities, where bacteria appear to be glycan biosynthesis, and synthesis of
sites. This shows the limitation of specialized in utilizing sugars associ- aromatic compounds, among others.
taxonomy-based studies, because the ated with the dentinary tissue and to If we focus on the red-complex
large redundancy and plasticity of degrade proteins, given rise to the species of periodontal disease, the
microbes allows different bacterial hypothesis that dental caries is not a above-mentioned functional studies
consortia to perform the same func- single disease but a tissue-dependent showed that all three species
tions (see e.g. Vaishampayan et al. process of different aetiologies undergo an activation of metallopro-
2010). Thus, some authors have (Simon-Soron et al. 2013). Among teases, peptidases and proteins
emphasized the importance of identi- the detected proteases, it was partic- involved in iron metabolism, sug-
fying what are oral microbes doing ularly relevant the identification of gesting that these may represent
rather than what taxonomic groups bacterial collagenases (as well as important virulence factors in these
are present at a given time (Belda- other proteases like glycoproteases, well-established oral pathogens.
Ferre et al. 2012, Duran-Pinedo & serine-proteases, carboxy-terminal However, those studies showed, sur-
Frias-Lopez 2015, Takahashi 2015). proteases and metalloproteases) sug- prisingly, that most upregulated vir-
Also, presence of a given organism gesting that, in addition to human ulence factors in periodontitis came
does not imply that it is metaboli- metalloproteinases (Tj aderhane et al. from organisms which are generally
cally active. An example is given by 1998), this large microbial-encoded not considered major periodontal
some bacteria which were present at proteolytic potential may signifi- pathogens (Duran-Pinedo et al.
low levels in 24-h supragingival den- cantly contribute to dentinal protein 2014b) in agreement with the idea
tal plaque, like Corynebacterium, but degradation (Takahashi & Nyvad, that the whole microbial community
which was found to be among the 2016, although experimental evi- could be providing the virulence out-
most active in the community dence with dentine isolates should put (Berezow & Darveau 2011).

2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
S34 Mira et al.

Microbial Community Structure as a


sufficient to recapitulate both the dys- Belda-Ferre, P., Alcaraz, L. D., Cabrera-Rubio,
R., Romero, H., Sim on-Soro, A., Pignatelli, M.
Cause versus a Consequence of
biosis and the disease phenotype. For
& Mira, A. (2012) The oral metagenome in
Caries and Periodontal Disease
example, transplantation experiments health and disease. Multidisciplinary Journal of
where the disease-associated gut Microbial Ecology 6, 4656.
To summarize, dysbiosis in caries and microbiota is transferred into healthy Belda-Ferre, P., Williamson, J., Sim 
on-Soro, A.,
periodontal disease may be defined as germ free animals have demonstrated Artacho, A., Jensen, O. N. & Mira, A. (2015)
The human oral metaproteome reveals poten-
deleterious perturbations to the that the disease phenotype can be tial biomarkers for caries disease. Proteomics
health-associated microbial popula- reproduced in the recipients in a vari- 15, 34973507.
tion structures of the oral microbiota ety of conditions including adiposity, Bentez-P
aez, A., Belda-Ferre, P., Simon-Soro, A.
and accompanying breakdown in the metabolic syndrome and colitis (Spor & Mira, A. (2014) Microbiota diversity and
gene expression dynamics in human oral bio-
normal homoeostatic balance et al. 2011). films. BMC Genomics 15, 311.
between the host and the resident Equally, the restoration of a nor- Berezow, A. B. & Darveau, R. P. (2011) Micro-
microbes. Multiple factors are likely mal healthy microbiota following bial shift and periodontitis. Periodontology
to be able to effect the perturbations. successful faecal transplantation for 2000 55, 3647.
Biesbroek, G., Sanders, E. A., Roeselers, G.,
In the case of caries, dietary sugar the treatment of Clostridium difficile Wang, X., Caspers, M. P., Trzci nski, K.,
and its metabolism by acid tolerant infection suggests that restoration of Bogaert, D. & Keijser, B. J. (2012) Deep
bacteria appears to have a profound a normal balanced microbiota in the sequencing analyses of low density microbial
influence on this dysbiosis and a gut is sufficient to restore intestinal communities: working at the boundary of accu-
direct linkage to the disease process. rate microbiota detection. PLoS ONE 7,
health (van Nood et al. 2013). Fae-
e32942.
In the case of periodontal dis- cal transplantation treatment of the Blanc, V., OValle, F., Pozo, E., Puertas, A.,
eases, apart from the specific role of other major inflammatory bowel dis- Le on, R. & Mesa, F. (2015) Oral bacteria in
the JP2 clone of A. actinomycetem- eases, ulcerative colitis and Crohns placental tissues: increased molecular detection
comitans in cases of aggressive peri- disease has so far yielded less suc- in pregnant periodontitis patients. Oral Dis-
eases 21, 905912.
odontitis (Haubek et al. 2008), the cessful outcomes. However, the prin- Bradshaw, D. J., Homer, K. A., Marsh, P. D. &
situation is less clear that dysbiosis is ciple of treating inflammatory Beighton, D. (1994) Metabolic cooperation in
causative or consequential of the dis- diseases associated with a perturbed oral microbial communities during growth on
ease. It is feasible to argue that alter- microbiome by modalities aimed mucin. Microbiology 140, 34073412.
Brailsford, S., Sheehy, E., Gilbert, S., Clark, D.
ations to the inflammatory status of entirely at conversion of the dysbi- C., Kidd, E. A., Zoitopoulos, L., Adams, S. E.,
a site will lead to the selection of otic microbiota to the previously Visser, J. M. & Beighton, D. (2005) The micro-
those members of the microbial com- harmonious and balanced system is flora of the erupting first permanent molar.
munity most well adapted to survive now firmly established. Such an Caries Research 39, 7884.
Byrne, D. P., Potempa, J., Olczak, T. & Smalley,
the increased inflammatory pressure approach may have some benefit in J.W. (2013) Evidence of mutualism between
and/or take maximal advantage of the treatment of periodontal disease two periodontal pathogens. Molecular Oral
the altered nutritional environment (Pozhitkov et al. 2015). Microbiology 28, 219229.
presented by such an alteration. Bac- Cabrera-Rubio, R., Collado, M. C., Laitinen, K.,
teria less well adapted to the injuri- Salminen, S., Isolauri, E. & Mira, A. (2012)
References The human milk microbiome changes over lac-
ous properties of the innate response tation and is shaped by maternal weight and
or ill-equipped to compete metaboli- Aagaard, K., Ma, J., Antony, K. M., Ganu, R., mode of delivery. American Journal of Clinical
cally in this new environment will Petrosino, J. & Versalovic, J. (2014) The pla- Nutrition 96, 544551.
diminish in their overall proportions centa harbors a unique microbiome. Science Camelo-Castillo, A., Bentez-P aez, A., Belda-
Translational Medicine 6, 237ra265. Ferre, P., Cabrera-Rubio, R. & Mira, A.
in the population. In this scenario, Aas, J. A., Griffen, A. L., Dardis, S. R., Lee, A. (2014) Streptococcus dentisani sp. nov., a novel
the dysbiosis is consequential not M., Olsen, I., Dewhirst, F. E., Leys, E. J. & member of the mitis group. International Jour-
causative. Further evidence that the Paster, B. J. (2008) Bacteria of dental caries in nal Systematic Evolutionary Microbiology 64,
environmental changes induced by primary and permanent teeth in children and 6065.
young adults. Journal of Clinical Microbiology Cameron, S. J., Huws, S. A., Hegarty, M. J.,
the diseased status of the host drive Smith, D. P. & Mur, L. A. (2015) The human
46, 14071417.
and sustain the alterations to the Aas, J. A., Paster, B. J., Stokes, L. N., Olsen, I. salivary microbiome exhibits temporal stability
microbiota rather than the reverse & Dewhirst, F. E. (2005) Defining the normal in bacterial diversity. FEMS Microbiology Ecol-
comes from the efficacy, in certain bacterial flora of the oral cavity. Journal of ogy 91, fiv091.
Clinical Microbiology 43, 57215732. Caporaso, J. G., Lauber, C. L., Costello, E. K.,
instances, of immuno-modulatory 
Aberg, C. H., Kelk, P. & Johansson, A. (2015) Berg-Lyons, D., Gonzalez, A., Stombaugh, J.,
drugs which target the inflammatory Aggregatibacter actinomycetemcomitans: viru- Knights, D., Gajer, P., Ravel, J., Fierer, N.,
systems of the host as opposed to lence of its leukotoxin and association with Gordon, J. I. & Knight, R. (2011) Moving pic-
direct intervention at the level of the aggressive periodontitis. Virulence 6, 188195. tures of the human microbiome. Genome Biol-
microbiota (Delima et al. 2002). Amann, R. I., Ludwig, W. & Schleifer, K. H. ogy 12, R50.
(1995) Phylogenetic identification and in situ Caufield, P. W., Cutter, G. R. & Dasanayake, A.
In other fields, there is now accu- detection of individual microbial cells without P. (1993) Initial acquisition of mutans strepto-
mulating evidence to support the cultivation. Microbiolical Reviews 59, 143169. cocci by infants: evidence for a discrete window
counter argument that dysbiosis in Babaahmady, K. G., Marsh, P. D., Challacombe, of infectivity. Journal of Dental Research 72,
itself is sufficient to both initiate and S. J. & Newman, H. N. (1997) Variations in 3745.
the predominant cultivable microflora of dental Cephas, K. D., Kim, J., Mathai, R. A., Barry, K.
drive disease. Perhaps the most com- plaque at defined subsites on approximal tooth A., Dowd, S. E., Meline, B. S. & Swanson, K.
pelling data has come from studies in surfaces in children. Archives of Oral Biology S. (2011) Comparative analysis of salivary bac-
animals over the last decade which 42, 101111. terial microbiome diversity in edentulous
have demonstrated that transfer of a Badet, C. & Thebaud, N. B. (2008) Ecology of infants and their mothers or primary care
lactobacilli in the oral cavity: a review of litera- givers using pyrosequencing. PLoS ONE 6,
dysbiotic microbiome from a diseased e23503.
ture. Open Microbiology Journal 2, 3848.
animal into a healthy recipient can be
2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Microbial communities S35

Chang, J. Y., Antonopoulos, D. A., Kalra, A., acquisition and structure of the initial micro- Gross, E. L., Beall, C. J., Kutsch, S. R., Fire-
Tonelli, A., Khalife, W. T., Schmidt, T. M. & biota across multiple body habitats in new- stone, N. D., Leys, E. J. & Griffen, A. L.
Young, V. B. (2008) Decreased diversity of the borns. Proceedings of the National Academy of (2012) Beyond Streptococcus mutans: dental
fecal Microbiome in recurrent Clostridium diffi- Sciences of the United States of America 107, caries onset linked to multiple species by 16S
cile-associated diarrhea. Journal of Infectious 1197111975. rRNA community analysis. PLoS ONE 7,
Diseases 197, 435438. Dominguez-Bello, M. G., De Jesus-Laboy, K. M., e47722.
Chen, L., Qin, B., Du, M., Zhong, H., Xu, Q., Shen, N., Cox, L. M., Amir, A., Gonzalez, A., Haffajee, A. D., Teles, R. P., Patel, M. R., Song,
Li, Y., Zhang, P. & Fan, M. (2015) Extensive Bokulich, N. A., Song, S. J., Hoashi, M., Riv- X., Yaskell, T. & Socransky, S. S. (2009) Fac-
description and comparison of human supra- era-Vinas, J. I., Mendez, K., Knight, R. & Cle- tors affecting human supragingival biofilm
gingival microbiome in root caries and health. mente, J. C. (2016) Partial restoration of the composition. II. Tooth position. Journal of
PLoS ONE 10, e0117064. microbiota of cesarean-born infants via vaginal Periodontal Research 44, 520528.
Clerehugh, V. & Tugnait, A. (2001) Periodontal microbial transfer. Nature Medicine 22, 250 Hajishengallis, G., Darveau, R. P. & Curtis, M.
diseases in children and adolescents: 1. Aetiol- 253. A. (2012) The keystone-pathogen hypothesis.
ogy and diagnosis. Dental Update 28, 222230, Duran-Pinedo, A. E., Baker, V. D. & Frias- Nature Reviews Microbiology 10, 717725.
232. Lopez, J. (2014a) The periodontal pathogen Hajishengallis, G., Liang, S., Payne, M. A.,
Collado, M. C., Rautava, S., Aakko, J., Isolauri, Porphyromonas gingivalis induces expression of Hashim, A., Jotwani, R., Eskan, M. A., McIn-
E. & Salminen, S. (2016) Human gut colonisa- transposases and cell death of Streptococcus tosh, M. L., Alsam, A., Kirkwood, K. L.,
tion may be initiated in utero by distinct micro- mitis in a biofilm model. Infection and Immu- Lambris, J. D., Darveau, R. P. & Curtis, M.
bial communities in the placenta and amniotic nity 82, 33743382. A. (2011) Low-abundance biofilm species
fluid. Scientific Reports 6, 23129. Duran-Pinedo, A. E., Chen, T., Teles, R., Starr, orchestrates inflammatory periodontal disease
Colombo, A. P., Boches, S. K., Cotton, S. L., J. R., Wang, X., Krishnan, K. & Frias-Lopez, through the commensal microbiota and com-
Goodson, J. M., Kent, R., Haffajee, A. D., J. (2014b) Community-wide transcriptome of plement. Cell Host & Microbe 10, 497506.
et al. (2009) Comparisons of subgingival micro- the oral microbiome in subjects with and with- Hashimoto, K., Sato, T., Shimauchi, H. & Taka-
bial profiles of refractory periodontitis, severe out periodontitis. Multidisciplinary Journal of hashi, N. (2011) Profiling of dental plaque
periodontitis, and periodontal health using the Microbial Ecology 8, 16591672. microflora on root caries lesions and the pro-
human oral microbe identification microarray. Duran-Pinedo, A. E. & Frias-Lopez, J. (2015) tein-denaturing activity of these bacteria. Amer-
Journal of Periodontology 80, 14211432. Beyond microbial community composition: ican Journal of Dentistry 24, 295299.
Costello, E. K., Lauber, C. L., Hamady, M., functional activities of the oral microbiome in Haubek, D., Ennibi, O. K., Poulsen, K., Vaeth,
Fierer, N., Gordon, J. I. & Knight, R. (2009) health and disease. Microbes and Infection 17, M., Poulsen, S. & Kilian, M. (2008) Risk of
Bacterial community variation in human body 505516. aggressive periodontitis in adolescent carriers
habitats across space and time. Science 326, Ebersole, J. L., Feuille, F., Kesavalu, L. & Holt, of the JP2 clone of Aggregatibacter (Acti-
16941697. S. C. (1997) Host modulation of tissue destruc- nobacillus) actinomycetemcomitans in Morocco:
Crielaard, W., Zaura, E., Schuller, A. A., Huse, tion caused by periodontopathogens: effects on a prospective longitudinal cohort study. Lancet
S. M., Montijn, R. C. & Keijser, B. J. (2011) a mixed microbial infection composed of Por- 2371, 237242.
Exploring the oral microbiota of children at phyromonas gingivalis and Fusobacterium nucle- Hold, G. L., Smith, M., Grange, C., Watt, E. R.,
various developmental stages of their dentition atum. Microbial Pathogenesis 23, 2332. El-Omar, E. M. & Mukhopadhya, I. (2014)
in the relation to their oral health. BMC Medi- Emilson, C. G. & Krasse, B. (1985) Support for Role of the gut microbiota in inflammatory
cal Genomics 4, 22. and implications of the specific plaque hypothe- bowel disease pathogenesis: what have we
Curtis, M. A. (2014) Periodontal microbiology sis. Scandinavian Journal of Dental Research 93, learnt in the past 10 years? World Journal of
the lids off the box again. Journal of Dental 96104. Gastroenterology 20, 11921210.
Research 93, 840842. Fardini, Y., Chung, P., Dumm, R., Joshi, N. & Holgerson, P. L., Harnevik, L., Hernell, O.,
Darveau, R. P., Belton, C. M., Reife, R. A. & Han, Y. W. (2010) Transmission of diverse oral Tanner, A. C. R. & Johansson, I. (2011)
Lamont, R. J. (1998) Local chemokine paraly- bacteria to murine placenta: evidence for the Mode of birth delivery affects oral microbiota
sis, a novel pathogenic mechanism for Porphy- oral microbiome as a potential source of in infants. Journal of Dental Research 90,
romonas gingivalis. Infection and Immunity 66, intrauterine infection. Infection and Immunity 11831188.
16601665. 78, 17891796. Holgerson, P. L., Vestman, N. R., Claesson, R.,
Darveau, R. P., Hajishengallis, G. & Curtis, M. Fejerskov, O. (2004) Changing paradigms in con- Ohman, C., Domell of, M., Tanner, A. C., Her-
A. (2012) Porphyromonas gingivalis as a poten- cepts on dental caries: consequences for oral nell, O. & Johansson, I. (2013) Oral microbial
tial community activist for disease. Journal of health care. Caries Research 38, 182191. profile discriminates breast-fed from formula-
Dental Research 91, 816820. Fejerskov, O., Nyvad, B. & Larsen, M.J. (1994) fed infants. Journal of Pediatric Gastroenterol-
David, L. A., Materna, A. C., Friedman, J., Cam- Human experimental caries models: intra-oral ogy and Nutrition 56, 127136.
pos-Baptista, M. I., Blackburn, M. C., Per- environmental variability. Advances in Dental
Holgerson, P.L., Ohman, C., R onnlund, A. &
rotta, A., Erdman, S. E. & Alm, E. J. (2014) Research 8, 134143. Johansson, I. (2015) Maturation of oral micro-
Host lifestyle affects human microbiota on Feuille, F., Ebersole, J. L., Kesavalu, L., Stepfen, biota in children with or without dental caries.
daily timescales. Genome Biology 15, R89. M. J. & Holt, S. C. (1996) Mixed infection PLoS One 10, e0128534.
Delima, A. J., Karatzas, S., Amar, S. & Graves, with Porphyromonas gingivalis and Fusobac- Huang, X., Palmer, S. R., Ahn, S. J., Richards,
D. T. (2002) Inflammation and tissue loss terium nucleatum in a murine lesion model: V. P., Williams, M. L., Nascimento, M. M. &
caused by periodontal pathogens is reduced by potential synergistic effects on virulence. Infec- Burne, R. A. (2016) A highly arginolytic Strep-
interleukin-1 antagonists. Journal of Infectious tion and Immunity 64, 20942100. tococcus species that potently antagonizes
Diseases 186, 511516. Frias-Lopez, J. & Duran-Pinedo, A. (2012) Effect Streptococcus mutans. Applied Environmental
Dewhirst, F. E., Chen, T., Izard, J., Paster, B. J., of periodontal pathogens on the metatranscrip- Microbiology 82, 21872201.
Tanner, A. C., Yu, W. H., Lakshmanan, A. & tome of a healthy multispecies biofilm model. Huang, S., Yang, F., Zeng, X., Chen, J., Li, R.,
Wade, W. G. (2010) The human oral micro- Journal of Bacteriology 194, 20822095. Wen, T., Li, C., Wei, W., Liu, J., Chen, L.,
biome. Journal of Bacteriology 192, 50025017. Ghannoum, M.A., Jurevic, R.J., Mukherjee, P.K., Davis, C. & Xu, J. (2011) Preliminary charac-
Dige, I., Grnkjr, L. & Nyvad, B. (2014) Cui, F., Sikaroodi, M., Naqvi, A. & Gillevet, terization of the oral microbiota of Chinese
Molecular studies of the structural ecology of P.M. (2010) Characterization of the oral fungal adults with and without gingivitis. BMC Oral
natural occlusal caries. Caries Research 48, microbiome (mycobiome) in healthy individu- Health 11, 33.
451460. als. PLoS Pathogens 6, e1000713. Hughes, C. V., Andersen, R. N. & Kolenbrander,
Do, T., Sheehy, E. C., Mulli, T., Hughes, F. & Gibbons, R. J. (1989) Bacterial adhesion to oral P. E. (1992) Characterization of Veillonella
Beighton, D. (2015) Transcriptomic analysis of tissues: a model for infectious diseases. Journal atypica PK1910 adhesin-mediated coaggrega-
three Veillonella spp. present in carious dentine of Dental Research 68, 750760. tion with oral Streptococcus spp. Infection and
and in the saliva of caries-free individuals. Griffen, A. L., Beall, C. J., Campbell, J. H., Fire- Immunity 60, 11781186.
Frontiers in Cellular and Infection Microbiology stone, N. D., Kumar, P. S., Yang, Z. K., Human Microbiome Project Consortium (2012)
5, 25. Podar, M. & Leys, E. J. (2012) Distinct and Structure, function and diversity of the healthy
Dominguez-Bello, M. G., Costello, E. K., Contr- complex bacterial profiles in human periodonti- human microbiome. Nature 486, 207214.
eras, M., Magris, M., Hidalgo, G., Fierer, N. tis and health revealed by 16S pyrosequencing. Huurre, A., Kalliomaki, M., Rautava, S., Rinne,
& Knight, R. (2010) Delivery mode shapes the ISME Journal 6, 11761185. M., Salminen, S. & Isolauri, E. (2008) Mode of

2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
S36 Mira et al.

deliveryeffects on gut microbiota and Lamont, R. J. & Hajishengallis, G. (2015) Polymi- Mira-Pascual, L., Cabrera-Rubio, R., Ocon, S.,
humoral immunity. Neonatology 93, 236240. crobial synergy and dysbiosis in inflammatory Costales, P., Parra, A., Suarez, A., Moris, F.,
Jeurink, P. V., van Bergenhenegouwen, J., disease. Trends in Molecular Medicine 21, 172 Rodrigo, L., Mira, A. & Collado, M. C. (2015)
Jimenez, E., Knippels, L. M., Fern andez, L., 183. Microbial mucosal colonic shifts associated
Garssen, J., Knol, J., Rodrguez, J. M. & Lauder, A. P., Roche, A. M., Sherrill-Mix, S., with the development of colorectal cancer
Martn, R. (2013) Human milk: a source of Bailey, A., Laughlin, A. L., Bittinger, K., Leite, reveal the presence of different bacterial and
more life than we imagine. Beneficial Microbes R., Elovitz, M. A., Parry, S. & Bushman, F. archaeal biomarkers. Journal of Gastroenterol-
4, 1730. D. (2016) Comparison of placenta samples with ogy 50, 167179.
Jiang, W., Ling, Z., Lin, X., Chen, Y., Zhang, J., contamination controls does not provide evi- Moore, W. E. C., Holdeman, L. V., Smibert, R.
Yu, J., Xiang, C. & Chen, H. (2014) Pyrose- dence for a distinct placenta microbiota. Micro- M., Hash, D. E., Burmeister, J. A. & Ranney,
quencing analysis of oral microbiota shifting in biome 4, 29. R. R. (1982) Bacteriology of severe periodonti-
various caries states in childhood. Microbial Law, V., Seow, W. K. & Townsend, G. (2007) tis in young adult humans. Infection and Immu-
Ecology 67, 962969. Factors influencing oral colonization of mutans nity 38, 11371148.
Jiang, W., Zhang, J. & Chen, H. (2013) Pyrose- streptococci in young children. Australian Den- Mor, G. & Kwon, J. Y. (2015) Trophoblast-
quencing analysis of oral microbiota in children tal Journal 52, 93100. microbiome interaction: a new paradigm on
with severe early childhood dental caries. Cur- Lepage, P., H asler, R., Spehlmann, M. E., Zvir- immune regulation. American Journal of
rent Microbiology 67, 537542. bliene, A., Begun, A., Ott, S., Kupcinskas, L., Obstetrics and Gynecology 213 (4 Suppl.),
Kesavalu, L., Sathishkumar, S., Bakthavatchalu, Dore, J., Raedler, A. & Schreiber, S. (2011) S131S137.
V., Matthews, C., Dawson, D., Steffen, M. & Twin study indicates loss of interaction Murray, J. L., Connell, J. L., Stacy, A., Turner,
Ebersole, J. L. (2007) Rat model of polymicro- between microbiota and mucosa of patients K. H. & Whiteley, M. (2014) Mechanisms of
bial infection, immunity, and alveolar bone with ulcerative colitis. Gastroenterology 141, synergy in polymicrobial infections. Journal of
resorption in periodontal disease. Infection and 227236. Microbiology 52, 188199.
Immunity 75, 17041712. Li, Y. & Caufield, P. W. (1995) The fidelity of ini- van Nood, E., Vrieze, A., Nieuwdorp, M., et al.
Kirst, M. E., Li, E. C., Alfant, B., Chi, Y. Y., tial acquisition of mutans streptococci by (2013) Duodenal infusion of donor feces for
Walker, C., Magnusson, I. & Wang, G. P. infants from their mothers. Journal Dental recurrent Clostridium difficile. New England
(2015) Dysbiosis and alterations in predicted Research 74, 681685. Journal of Medicine 368, 407415.
functions of the subgingival microbiome in Li, Y., Caufield, P. W., Dasanayake, A. P., Wiener, Noorda, W. D., Purdell-Lewis, D. J., van Mont-
chronic periodontitis. Applied and Environment H. W. & Vermund, S. H. (2005) Mode of deliv- fort, A. M. & Weerkamp, A. H. (1988)
Microbiology 81, 783793. ery and other maternal factors influence the Monobacterial and mixed bacterial plaques of
Kistler, J. O., Booth, V., Bradshaw, D. J. & acquisition of Streptococcus mutans in infants. Streptococcus mutans and Veillonella alcalescens
Wade, W. G. (2013) Bacterial community Journal of Dental Research 84, 806811. in an artificial mouth: development, metabo-
development in experimental gingivitis. PLoS Li, Y., Navia, J. M. & Caufield, P. W. (1994) lism, and effect on human dental enamel. Car-
ONE 8, e71227. Colonization by mutans streptococci in the ies Research 22, 342347.
Kleinberg, I. & Jenkins, G. N. (1964) The pH of mouths of 3- and 4-year-old Chinese children Nyvad, B., Crielaard, W., Mira, A., Takahashi,
dental plaques in the different areas of the with or without enamel hypoplasia. Archives of N. & Beighton, D. (2013) Dental caries from a
mouth before and after meals and their rela- Oral Biology 39, 10571062. molecular microbiological perspective. Caries
tionship to the pH and rate of flow of resting Ling, Z., Kong, J., Jia, P., Wei, C., Wang, Y., Research 47, 89102.
saliva. Archives of Oral Biology 9, 493516. Pan, Z., Huang, W., Li, L., Chen, H. & Xiang, Nyvad, B. & Fejerskov, O. (1997) Assessing the
Koga, K., Aldo, P. B. & Mor, G. (2009) Toll-like C. (2010) Analysis of oral microbiota in chil- stage of caries lesion activity on the basis of
receptors and pregnancy: trophoblast as modu- dren with dental caries by PCR-DGGE and clinical and microbiological examination. Com-
lators of the immune response. Journal of barcoded pyrosequencing. Microbial Ecology munity Dentistry and Oral Epidemiology 25, 69
Obstetrics and Gynaecology Research 35, 191 60, 677690. 75.
202. Loesche, W. J. (1986) Role of Streptococcus Nyvad, B. & Kilian, M. (1990) Microflora associ-
Kohler, B., Andreen, I. & Jonsson, B. (1988) The mutans in human dental decay. Microbiological ated with experimental root surface caries in
earlier the colonization by mutans streptococci, Reviews 50, 353380. humans. Infection and Immunity 58, 16281633.
the higher the caries prevalence at 4 years of Manichanh, C., Rigottier-Gois, L., Bonnaud, E., Obata, J., Takeshita, T., Shibata, Y., Yamanaka,
age. Oral Microbiology and Immunology 3, 14 Gloux, K., Pelletier, E., Frangeul, L., Nalin, W., Unemori, M., Akamine, A. & Yamashita,
17. R., Jarrin, C., Chardon, P., Marteau, P., Roca, Y. (2014) Identification of the microbiota in
Kolenbrander, P. E., Palmer, R. J. Jr, Rickard, J. & Dore, J. (2006) Reduced diversity of faecal carious dentin lesions using 16S rRNA gene
A. H., Jakubovics, N. S., Chalmers, N. I. & microbiota in Crohns disease revealed by a sequencing. PLoS ONE 9, e103712.
Diaz, P. I. (2006) Bacterial interactions and metagenomic approach. Gut 55, 205211. Oh, T. J., Eber, R. & Wang, H. L. (2002)
successions during plaque development. Peri- Mantzourani, M., Fenlon, M. & Beighton, D. Periodontal diseases in the child and adoles-
odontology 2000 42, 4779. (2009) Association between Bifidobacteriaceae cent. Journal of Clinical Periodontology 29,
Kon onen, E., Kanervo, A., Takala, A., Asikai- and the clinical severity of root caries lesions. 400410.
nen, S. & Jousimies-Somer, H. (1999) Estab- Oral Microbiology and Immunology 24, 3237. Perez-Chaparro, P. J., Goncalves, C., Figueiredo,
lishment of oral anaerobes during the first year Marsh, P. D. (2003) Are dental diseases examples L. C., Faveri, M., Lob~ ao, E., Tamashiro, N.,
of life. Journal of Dental Research 78, 1634 of ecological catastrophes? Microbiology 149, et al. (2014) Newly identified pathogens associ-
1639. 279294. ated with periodontitis: a systematic review.
Kon onen, E. & M uller, H. P. (2014) Microbiol- Marsh, P. D., Head, D. A. & Devine, D. A. Journal of Dental Research 93, 846858.
ogy of aggressive periodontitis. Periodontology (2015) Ecological approaches to oral biofilms: Peterson, S. N., Meissner, T., Su, A. I., Snesrud,
2000 65, 4678. control without killing. Caries Research 49 E., Ong, A. C., Schork, N. J. & Bretz, W. A.
Koopman, J. E., van der Kaaij, N. C., Buijs, M. (Suppl. 1), 4654. (2014) Functional expression of dental plaque
J., Elyassi, Y., van der Veen, M. H., Crielaard, Mikx, F. H. & van der Hoeven, J. S. (1975) Sym- microbiota. Frontiers in Cellular and Infection
W., Ten Cate, J. M. & Zaura, E. (2015) The biosis of Streptococcus mutans and Veillonella Microbiology 4, 108.
effect of fixed orthodontic appliances and fluo- alcalescens in mixed continuous cultures. Pham, T. A. & Lawley, T. D. (2014) Emerging
ride mouthwash on the oral microbiome of Archives of Oral Biology 20, 407410. insights on intestinal dysbiosis during bacterial
adolescents a randomized controlled clinical Milgrom, P., Riedy, C. A., Weinstein, P., Tanner, infections. Current Opinion in Microbiology 17,
trial. PLoS ONE 10, e0137318. A. C., Manibusan, L. & Bruss, J. (2000) Dental 6774.
Krom, B. P., Kidwai, S. & Ten Cate, J. M. (2014) caries and its relationship to bacterial infection, Pozhitkov, A. E., Leroux, B. G., Randolph, T.
Candida and other fungal species: forgotten hypoplasia, diet, and oral hygiene in 6- to 36- W., Beikler, T., Flemmig, T. F. & Noble, P. A.
players of healthy oral microbiota. Journal of month-old children. Community Dentistry and (2015) Towards microbiome transplant as a
Dental Research 93, 445451. Oral Epidemiology 28, 295306. therapy for periodontitis: an exploratory study
Lai, P. Y., Seow, W. K., Tudehope, D. I. & Mira, A. & Rodriguez, J. M. (2016) The origin of of periodontitis microbial signature contrasted
Rogers, Y. (1997) Enamel hypoplasia and den- human milk bacteria. Ch. 13. In: McGuire, M., by oral health, caries and edentulism. BMC
tal caries in very-low birthweight children: a McGuire, M. & Bode, L. (eds). Prebiotics and Oral Health 15, 125.
case-controlled, longitudinal study. Pediatric Probiotics in Human Breast Milk, pp. 301311. Prince, A. L., Ma, J., Kannan, P. S., Alvarez, M.,
Dentistry 19, 4249. London: Elsevier. Gisslen, T., Harris, R. A., Sweeney, E. L.,

2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Microbial communities S37

Knox, C. L., Lambers, D. S., Jobe, A. H., complexes in subgingival plaque. Journal of Egholm, M., Henrissat, B., Heath, A.C.,
Chougnet, C. A., Kallapur, S. G. & Aagaard, Clinical Periodontology 25, 134144. Knight, R. & Gordon, J.I. (2009) A core gut
K. M. (2016) The placental membrane micro- Spor, A., Koren, O. & Ley, R. (2011) Unravelling microbiome in obese and lean twins. Nature
biome is altered among subjects with sponta- the effects of the environment and host geno- 457, 480484.
neous preterm birth with and without type on the gut microbiome. Nature Reviews Utter, D. R., Mark Welch, J. L. & Borisy, G. G.
chorioamnionitis. American Journal of Obstet- Microbiology 9, 279290. (2016) Individuality, stability, and variability of
rics and Gynecology 214, 627. e1627.e16. Stacy, A., Everett, J., Jorth, P., Trivedi, U., Rum- the plaque microbiome. Frontiers in Microbiol-
Proceedings of the World Workshop (1996) Pro- baugh, K. P. & Whiteley, M. (2014) Bacterial ogy 22, 564.
ceedings of the 1996 World Workshop in Peri- fight-and-flight responses enhance virulence in Vaishampayan, P. A., Kuehl, J. V., Froula, J. L.,
odontics Lansdowne, Virginia, July 1317, a polymicrobial infection. Proceedings of the Morgan, J. L., Ochman, H. & Francino, M. P.
1996. Annals of Periodontology 1, 1947. National Academy of Sciences of the United (2010) Comparative metagenomics and popula-
Rotimi, V. O. & Duerden, B. I. (1981) The devel- States of America 111, 78197824. tion dynamics of the gut microbiota in mother
opment of the bacterial flora in normal Stahringer, S. S., Clemente, J. C., Corley, R. P., and infant. Genome Biology and Evolution 2,
neonates. Journal of Medical Microbiology 14, Hewitt, J., Knights, D., Walters, W. A., 5366.
5162. Knight, R. & Krauter, K. S. (2012) Nurture Verma, R., Bhattacharyya, I., Sevilla, A., Lieber-
Rutayisire, E., Huang, K., Liu, Y. & Tao, F. trumps nature in a longitudinal survey of sali- man, I., Pola, S., Nair, M., Wallet, S. M.,
(2016) The mode of delivery affects the vary bacterial communities in twins from early Aukhil, I. & Kesavalu, L. (2010b) Virulence of
diversity and colonization pattern of the gut adolescence to early adulthood. Genome major periodontal pathogens and lack of
microbiota during the first year of infants life: Research 22, 21462152. humoral immune protection in a rat model of
a systematic review. BMC Gastroenterology 16, Takahashi, N. (2015) Oral microbiome metabo- periodontal disease. Oral Diseases 16, 686695.
86. lism: from who are they? to what are they Verma, R. K., Rajapakse, S., Meka, A., Hamrick,
Sabatos-Peyton, C. A., Verhagen, J. & Wraith, D. doing?. Journal of Dental Research 94, 1628 C., Pola, S., Bhattacharyya, I., Nair, M., Wal-
C. (2010) Antigen-specific immunotherapy of 1637. let, S. M., Aukhil, I. & Kesavalu, L. (2010a)
autoimmune and allergic diseases. Current Takahashi, N. & Nyvad, B. (2011) The role of Porphyromonas gingivalis and Treponema denti-
Opinion in Immunology 22, 609615. bacteria in the caries process: ecological per- cola mixed microbial infection in a rat model
Segata, N., Haake, S. K., Mannon, P., Lemon, spectives. Journal of Dental Research 90, 294 of periodontal disease. Interdisciplinary Per-
K. P., Waldron, L., Gevers, D., Huttenhower, 303. spectives on Infectious Diseases 2010, 605125.
C. & Izard, J. (2012) Composition of the adult Takahashi, N. & Nyvad, B. (2016) Ecological Walker, A. W. & Lawley, T. D. (2013) Therapeu-
digestive tract bacterial microbiome based on hypothesis of dentin and root caries. Caries tic modulation of intestinal dysbiosis. Pharma-
seven mouth surfaces, tonsils, throat and stool Research 50, 422431. cological Research 69, 7586.
samples. Genome Biology 13, R42. Tan, K. H., et al. (2014) Porphyromonas gingivalis Wan, A. K., Seow, W. K., Walsh, L. J., Bird, P.,
Shaddox, L. M., Huang, H., Lin, T., Hou, W., and Treponema denticola exhibit metabolic Tudehope, D. L. & Purdie, D. M. (2001) Asso-
Harrison, P. L., Aukhil, I., Walker, C. B., Kle- symbioses. PLoS Pathogens 10, e1003955. ciation of Streptococcus mutans infection and
pac-Ceraj, V. & Paster, B. J. (2012) Microbio- Tanner, A. C., Mathney, J. M., Kent, R. L., oral developmental nodules in pre-dentate
logical characterization in children with Chalmers, N. I., Hughes, C. V., Loo, C. Y., infants. Journal of Dental Research 80, 1945
aggressive periodontitis. Journal of Dental Pradhan, N., Kanasi, E., Hwang, J., Dahlan, 1948.
Research 91, 927933. M. A., Papadopolou, E. & Dewhirst, F. E. Yost, S., Duran-Pinedo, A. E., Teles, R., Krish-
Shah, A. G., Shetty, P. C., Ramachandra, C. S., (2011) Cultivable anaerobic microbiota of sev- nan, K. & Frias-Lopez, J. (2015) Functional
Bhat, N. S. & Laxmikanth, S. M. (2011) In ere early childhood caries. Journal of Clinical signatures of oral dysbiosis during periodontitis
vitro assessment of photocatalytic titanium Microbiology 49, 14641474. progression revealed by microbial metatran-
oxide surface modified stainless steel orthodon- Teles, R., Teles, F., Frias-Lopez, J., Paster, B. & scriptome analysis. Genome Medicine 7, 27.
tic brackets for antiadherent and antibacterial Haffajee, A. (2013) Lessons learned and Zaura, E., Keijser, B. J., Huse, S. M. & Crielaard,
properties against Lactobacillus acidophilus. unlearned in periodontal microbiology. Peri- W. (2009) Defining the healthy core micro-
Angle Orthodontist 81, 10281035. odontology 2000 62, 95162. biome of oral microbial communities. BMC
Silva, N., Abusleme, L., Bravo, D., Dutzan, N., Teughels, W., Loozen, G. & Quirynen, M. (2011) Microbiology 9, 259.
Garcia-Sesnich, J., Vernal, R., Hern andez, M. Do probiotics offer opportunities to manipulate Zaura, E., Nicu, E. A., Krom, B. P. & Keijser, B.
& Gamonal, J. (2015) Host response mecha- the periodontal oral microbiota? Journal Clini- J. (2014) Acquiring and maintaining a normal
nisms in periodontal diseases. Journal of cal Periodontology 38 (Suppl. 11), 159177. oral microbiome: current perspective. Frontiers
Applied Oral Science 23, 329355. Theilade, E. (1986) The non-specific theory in in Cellular and Infection Microbiology 4, 85.
Sim on-Soro, A., Belda-Ferre, P., Cabrera-Rubio, microbial etiology of inflammatory periodontal Zhang, S. (2014) Dental caries and vaccination
R., Alcaraz, L. D. & Mira, A. (2012) A tissue- diseases. Journal of Clinical Periodontology 13, strategy against the major cariogenic pathogen,
dependent hypothesis of dental caries. Caries 905911. Streptococcus mutans. Current Pharmaceutical
Research 47, 591600. Tj
aderhane, L., Larjava, H., Sorsa, T., Uitto, V. Biotechnology 14, 960966.
Sim on-Soro, A., Guillen-Navarro, M. & Mira, A. J., Larmas, M. & Salo, T. (1998) The activa-
(2014) Metatranscriptomics reveals overall tion and function of host matrix metallopro-
active bacterial composition in caries lesions. teinases in dentin matrix breakdown in caries Address:
Journal of Oral Microbiology 6, 25443. lesions. Journal of Dental Research 77, 1622 Alex Mira
Sim on-Soro, A. & Mira, A. (2015) Solving the eti- 1629. Department of Health and Genomics
ology of dental caries. Trends in Microbiology Torlakovic, L., Klepac-Ceraj, V., Ogaard, B., Center for Advanced Research in Public
23, 7682. Cotton, S. L., Paster, B. J. & Olsen, I. (2012)
Health
Sim on-Soro, A., Tomas, I., Cabrera-Rubio, R., Microbial community succession on developing
FISABIO Foundation
Catalan, M. D., Nyvad, B. & Mira, A. (2013) lesions on human enamel. Journal of Oral
Microbial geography of the oral cavity. Journal Microbiology 4, 16125. Valencia
of Dental Research 92, 616621. Turnbaugh, P.J., Hamady, M., Yatsunenko, T., Spain
Socransky, S. S., Haffajee, A. D., Cugini, M. A., Cantarel, B.L., Duncan, A., Ley, R.E., Sogin, E-mail: mira_ale@gva.es
Smith, C. & Kent, R. L. (1998) Microbial M.L., Jones, W.J., Roe, B.A., Affourtit, J.P.,

2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
S38 Mira et al.

Clinical Relevance individuals. There are highly signifi- Practical implications: An


Scientific rationale: Dental plaque cant differences in the microbiology, improved understanding of the
is composed of a complex mixture both between individuals and mechanisms behind the composi-
of different bacterial species which between sites in the same individual tional changes in the microbial
appear to be organized into an but an overall temporal stability at a communities of the mouth could
organized community structure given environmental site. Oral enable the development of preven-
with the potential to have an pathogens appear to be present at tative approaches and/or methods
impact on the maintenance of low frequencies in healthy individu- to reverse disease-associated
health and the development of als. However, stressors on these nor- microbial community structures to
disease. mally stable systems can lead to their status in health, although
Principal findings: Review of the lit- dramatic compositional and func- specific antimicrobial strategies
erature indicated that the directed tional changes in the microbiome (i.e. passive or active immuniza-
acquisition of the oral microbial and harmful effects to the oral tis- tion) may not be effective.
communities may vary between sues.

2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

Das könnte Ihnen auch gefallen