You are on page 1of 11

J Clin Periodontol 2017; 44 (Suppl. 18): S12–S22 doi: 10.1111/jcpe.

12679

Dental biofilm: ecological P. D. Marsh1 and Egija Zaura2


1
Department of Oral Biology, School of
Dentistry, University of Leeds, Leeds, UK;

interactions in health and 2


Department of Preventive Dentistry,
Academic Centre for Dentistry Amsterdam,
University of Amsterdam and Vrije

disease Universiteit, Amsterdam, The Netherlands

Marsh PD, Zaura E. Dental biofilm: ecological interactions in health and disease. J
Clin Periodontol 2017; 44 (Suppl. 18): S12–S22. doi: 10.1111/jcpe.12679.

Abstract
Background: The oral microbiome is diverse and exists as multispecies microbial
communities on oral surfaces in structurally and functionally organized biofilms.
Aim: To describe the network of microbial interactions (both synergistic and
antagonistic) occurring within these biofilms and assess their role in oral health
and dental disease.
Methods: PubMed database was searched for studies on microbial ecological
interactions in dental biofilms. The search results did not lend themselves to sys-
tematic review and have been summarized in a narrative review instead.
Results: Five hundred and forty-seven original research articles and 212 reviews
were identified. The majority (86%) of research articles addressed bacterial–bacte-
rial interactions, while inter-kingdom microbial interactions were the least stud-
ied. The interactions included physical and nutritional synergistic associations,
antagonism, cell-to-cell communication and gene transfer.
Conclusions: Oral microbial communities display emergent properties that cannot
be inferred from studies of single species. Individual organisms grow in environ-
ments they would not tolerate in pure culture. The networks of multiple synergis-
tic and antagonistic interactions generate microbial inter-dependencies and give
Key words: ecology; gene transfer;
biofilms a resilience to minor environmental perturbations, and this contributes interactions; metabolism; oral microbiome;
to oral health. If key environmental pressures exceed thresholds associated with signalling
health, then the competitiveness among oral microorganisms is altered and dys-
biosis can occur, increasing the risk of dental disease. Accepted for publication 19 December 2016

The mouth supports the growth of microbiome, which generally exists in microbiome and the host is symbiotic
diverse communities of microorgan- harmony with the host, and delivers or potentially damaging (dysbiotic),
isms – viruses, mycoplasmas, bacte- important benefits that contribute to thereby increasing the risk of diseases
ria, Archaea, fungi and protozoa overall health and well-being. The such as caries or periodontal diseases
(Wade 2013). These communities per- microorganisms found within these (Marsh 2003, Roberts & Darveau
sist on all surfaces as multispecies bio- oral biofilms live in close proximity 2015). Our aim was to review system-
films and form the resident oral with one another, which results in a atically the literature on microbial
wide range of potential interactions, interactions in dental biofilms in
which can be synergistic or antagonis- health and disease. However, the
Conflict of interest and source of tic. The composition of the micro- search strategy and outcomes, pre-
funding statement biome is influenced by the oral sented below, led to a conclusion that
The authors have stated explicitly
environment, and changes in local the topic is too broad for a systematic
that there are no conflict of interests
conditions can affect the microbial report and so the results are presented
in connection with this article.
No funding has been available other
interactions within these oral commu- as a narrative review, highlighting the
than that of the author’s institution.
nities and determine, in part, whether main microbial interactions in dental
the relationship between the oral biofilms in health and introducing the

S12 © 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Oral microbial interactions S13

environmental drivers for ecological as production of bacteriocins and listed in Table 2 and can be synergis-
dysbiosis towards disease. other inhibitory substances, cell-to- tic or antagonistic to the participat-
cell communication and gene transfer. ing species (Hojo et al. 2009, Huang
The bacterial species involved ranged et al. 2011, Kolenbrander 2011, Diaz
Literature search
from primary colonizers to taxa asso- 2012, Guo et al. 2014, Jakubovics
A PubMed search procedure was ciated with caries and periodontal dis- 2015a, Nobbs & Jenkinson 2015, Ng
performed on 19-07-2016. The query ease. Only 45 (8.2%) of the studies et al. 2016).
combined four separate search items: involved fungi, while interactions
(i) “microbiota,” including either involving viruses (18 studies), Archaea
Synergistic interactions
bacteria, viruses, Archaea, fungi, (four studies) and protozoa (three
protozoa or mycoplasma; (ii) “oral,” studies) were the least studied. Inter-
Physical interactions and biofilm
including distinct oral niches; (iii) kingdom interactions were addressed architecture
interactions, including either “ecol- in 71 studies, with the majority of
ogy,” “interaction,” “synergy,” “in- these focusing on Candida albicans Oral microorganisms must attach
hibition,” “co-occurrence,” and oral streptococci (Table 1). to surfaces if they are to persist in
“communication,” “metabolism,” Due to the high number of arti- the mouth and avoid being lost
“nutrients,” “gene transfer” or “quo- cles included and the broad range in by swallowing. Evidence primarily
rum sensing”; and (iv) “plaque,” the methods and the outcomes derived from laboratory studies sug-
“biofilm,” “community” or “consor- among the studies found, it was not gests that early colonizers adhere via
tium” (Table S1). This resulted in possible to report on the results in specific adhesin-receptor mechanisms
3758 hits. Of these, 3593 passed the the form of a systematic review or to molecules in the conditioning films
English language filter. After the meta-analysis. Instead, the articles that coat oral surfaces (Hojo et al.
screening of the titles and abstracts, that were identified by the described 2009), although, ultimately, microbial
the entries that did not relate to the search procedure were used as the growth is the major contributor to the
topic were excluded, leaving 759 arti- basis of the narrative review below. increase in biofilm biomass (Dige
cles. Among these were 212 reviews. et al. 2007). Oral microorganisms
The vast majority (86%) of the have a natural tendency to adhere to
Microbial interactions in health
original research articles (N = 547) other microbes and this process (co-
addressed bacterial interactions The close physical proximity of adhesion – the adherence of plank-
(Table 1). These included physical microorganisms within oral biofilms tonic cells to already attached organ-
(e.g. co-aggregation, co-adhesion) inevitably increases the probability isms on a surface) facilitates the
and nutritional synergistic interac- of interactions occurring. The most formation of multispecies biofilms
tions, antagonistic interactions such common types of interaction are (Kolenbrander 2011). In addition to
anchoring a cell to a surface, co-adhe-
sion also promotes microbial interac-
tions by co-locating organisms next to
Table 1. Details on original research articles (N = 547) obtained in PubMed search physiologically relevant partner spe-
described in Table S1 cies, thereby facilitating nutritional
Members of the Details co-operation and food chains, gene
interaction(s) transfer and cell–cell signalling. Sub-
stantial changes in gene expression
Bacteria–Bacteria Oral health: N = 205; caries pathogen (CP): N = 107;
occur when cells are in close proxim-
(N = 473) periodontal pathogen (PP): N = 149; CP and PP: N = 6;
Oral versus non-oral species: N = 6
ity or physical contact with one
Antagonism (A): N = 116; synergy (S): N = 214; A and S: N = 3; another (Wright et al. 2013), while
metabolism: N = 98; communication: N = 32; gene transfer: N = 10 functional consequences can result,
Bacteria–Fungi Candida albicans: N = 40; C. albicans and other Candida species: such as the protection of obligately
(N = 45) N = 3; undefined Candida spp.: N = 2 anaerobic bacteria in aerobic environ-
Bacteria involved: Streptococcus mutans: N = 11; ments by neighbouring species that
Streptococcus gordonii: N = 10; other oral streptococci: N = 9; either consume oxygen (Bradshaw
Actinomyces: N = 5; et al. 1994) or are oxygen-tolerating
Staphylococcus aureus N = 2; Aggregatibacter actinomycetemcomitans, (Diaz et al. 2002). Candida albicans
Enterococcus faecalis, Fusobacterium nucleatum N = 1 each; probiotic
can also co-aggregate with oral strep-
lactobacilli: N = 1; microbial consortia or microcosm: N = 8
Antagonism: N = 11; synergy: N = 33; communication: N = 5 tococci and can form synergistic part-
Bacteria–Viruses Bacteriophages: N = 6; Herpes viruses: N = 7; virome: N = 3; nerships in which the yeast promotes
(N = 18) CRISPR: N = 3 streptococcal biofilm formation, while
Bacteria–Archaea Methanogenic Archaea and periodontal pathogens streptococci enhance the invasive
(N = 4) property of Candida (Diaz et al. 2012,
Fungi–Fungi Different Candida species: N = 6; Pichia versus opportunistic fungi Xu et al. 2014). These physical and
(N = 7) functional associations can manifest
Fungi–Viruses HSV enhances C. albicans adherence themselves in some of the complex
(N = 1)
multispecies arrangements observed
Bacteria–Protozoa Bdellovibrio bacteriovorus – bacterial predator
(N = 3)
in oral biofilms formed in vivo, such
as “corn cob,” “test-tube brush” and
© 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
S14 Marsh and Zaura

Table 2. Types of synergistic and antagonistic microbial interactions that occur among oral biofilms in order to enable them to
microorganisms growing in dental plaque biofilms degrade host proteins and glycopro-
Interactions teins as nutrient sources (ter Steeg
Synergistic Antagonistic et al. 1987, ter Steeg & van der Hoe-
ven 1989). These interactions are dis-
Enzyme complementation/enzyme Bacteriocin production cussed in more detail later in the
sharing section on “Ecological drivers
Food chains (food webs) Hydrogen peroxide production
towards dysbiosis and disease”.
Co-adhesion Organic acid production/generation of inhibitory
pH conditions
Nutritional inter-dependencies
Cell–cell signalling Bacteriophage release such as those described above con-
Gene transfer Competition for essential nutrients tribute to the temporal stability and
Environmental modification Predation resilience of oral microbial communi-
ties, while a consequence of the reli-
ance of resident oral bacteria on the
metabolism of these complex sub-
“hedgehog” structures (Zijnge et al. Burne 2001, Koo et al. 2013). Indi-
2010, Dige et al. 2014, Mark Welch vidual bacteria are dependent there- strates is that species avoid direct
competition for individual nutrients,
et al. 2016). fore on the metabolic capability of
other species for access to essential and hence are able to co-exist and
maintain a stable equilibrium, also
nutrients.
Nutritional interactions
Further complex nutritional inter- termed microbial homeostasis
The primary nutrients for oral (Alexander 1971, Marsh 1989). This
relationships develop in microbial
microorganisms are host proteins and communities when the products of has been elegantly demonstrated in a
glycoproteins, and these are obtained computational study on KEGG path-
metabolism of one organism (pri-
mainly from saliva for organisms in mary feeder) become the main way-based metabolic distances
supragingival plaque (for a review, between 11 oral bacteria that are
source of nutrients for another (sec-
see Jakubovics 2015b) and from gin- ondary feeder), resulting in the known to interact (Mazumdar et al.
gival crevicular fluid (GCF) for those 2013). Metabolism was a major factor
development of food chains or food
located in subgingival biofilms (Wei webs (Hojo et al. 2009) (some exam- driving the order of colonization,
et al. 1999). Pure cultures of oral with specific metabolic pathways
ples are illustrated in Fig. 1). These
microorganisms grow poorly or not food webs can result in the complete associated with different layers in the
at all on these structurally complex biofilm, resulting in a functionally
and energetically efficient catabolism
substrates, and consortia of interact- of complex host molecules to the structured community. However, in
ing species are needed for their cata- simplest end products of metabolism such a structured community, there
bolism. Proteins are broken down by (e.g. CO2, CH4, H2S). Numerous was an optimal trade-off between
the action of mixtures of proteases synergistic metabolic interactions their resource sharing and functional
and peptidases, but the catabolism of occur among bacteria in subgingival synergy (Mazumdar et al. 2013).
glycoproteins (consisting of a protein
backbone decorated with linear or
branched oligosaccharide side chains)
involves the sequential removal of ter-
minal sugars from side chains before
the protein backbone becomes acces-
sible to proteolytic attack (Takahashi
2015). Oral bacteria express glycosi-
dases with different specificities so
that the concerted action of several
species is necessary for the complete
degradation of host glycoproteins
(Bradshaw et al. 1994). Similarly,
combinations of mutans streptococci,
Streptococcus oralis and Fusobac-
terium nucleatum, degraded albumin
more effectively than any of the three
species alone (Homer & Beighton
1992). The biofilm matrix is another
potential source for carbon and
energy for interacting consortia of
oral bacteria. Fructans and soluble
glucans in dental plaque can be
metabolized by combinations of bac-
teria that produce exo- and/or endo- Fig. 1. Examples of nutritional interactions among oral microorganisms (figure
hydrolytic enzymes (Bergeron & modified from Hojo et al. 2009).

© 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Oral microbial interactions S15

Cell–cell signalling dependent on the concentration of Porphyromonas gingivalis and F. nu-


Laboratory studies have shown that the signalling molecules. These cell– cleatum (Ali Mohammed et al.
microbial cells are able to communi- cell signalling strategies could enable 2013).
cate with, and respond to, neigh- cells to sense and adapt to various Evidence for HGT in dental bio-
bouring cells in biofilms by means of environmental stresses and, thereby, films has come from the discovery
small, diffusible, effector molecules. regulate (and coordinate) the expres- that both resident (S. mitis, S. oralis)
Gram-positive cells produce peptides sion of genes that influence the abil- and pathogenic (S. pneumoniae) bac-
that generally have a narrow spec- ity of pathogens to cause disease. teria isolated from the naso-pharyn-
trum of activity. In Streptococcus geal area possess genes conferring
mutans, two peptides [competence- Gene transfer
penicillin resistance that display a
stimulating peptide (CSP) and sigma common mosaic structure (Chi et al.
X-inducing peptide] promote genetic The close proximity of cells in bio- 2007). Similar evidence suggests
competence in other cells of S. mu- films provides ideal conditions for sharing of genes encoding for peni-
tans; production of these peptides is horizontal gene transfer (HGT). cillin-binding proteins among resi-
influenced by the local pH (Guo HGT involves either acquisition of dent oral and pathogenic Neisseria
et al. 2014) and carbohydrate source DNA from co-resident species or species (Bowler et al. 1994), and IgA
(Moye et al. 2014). CSP-mediated from exogenous sources (Petersen protease encoding genes among a
quorum sensing has also been identi- et al. 2005, Roberts & Kreth 2014). range of oral streptococcal species
fied in Streptococcus gordonii and DNA can be transferred through: (Poulsen et al. 1998).
Streptococcus intermedius. The func- transduction by bacterial viruses
tion of CSPs is to alter gene tran- (bacteriophages), conjugation by
Antagonistic interactions
scription and protein synthesis bacterial pili and transformation by
involved in biofilm formation, com- DNA uptake involving naturally A considerable number of studies
petence development, bacteriocin competent bacteria; in addition to addressed antagonistic interactions
synthesis, stress resistance, and the mechanisms above, DNA can involving inter-species and inter-
autolysis (Senadheera & Cvitkovitch also be transferred via membrane kingdom competition or “warfare”.
2008, Guo et al. 2014). Some strep- vesicles in Gram-negative bacteria The production of antagonistic com-
tococci can inactivate CSPs and (Olsen et al. 2013). HGT allows oral pounds such as bacteriocins, hydro-
thereby inhibit biofilm formation by bacteria to sample from an immense gen peroxide, organic acids, different
S. mutans (Wang et al. 2011). CSP metagenome and in this way increase enzymes and release of lytic phages
produced by S. gordonii can also their adaptive potential to changes are just a few examples of “weap-
inhibit biofilm formation by C. albi- in the oral environment (Roberts & ons” that can give an organism a
cans (Jack et al. 2015), so it is possi- Kreth 2014). For instance, metabolic competitive advantage during colo-
ble that a complex network of adaptability to carbohydrate-rich nization and when competing with
signalling interactions will exist in a environments such as the oral cavity other microbes (Table 2).
multispecies biofilm such as dental and gut has been found in a Lacto- Bacteriocins and bacteriocin-like
plaque. bacillus salivarius strain carrying a substances are produced by both
Autoinducer-2 (AI-2) is produced plasmid with genes involved in gly- Gram-positive and Gram-negative
by several genera of oral Gram-posi- colysis (Roberts & Kreth 2014). bacteria, with the most studied oral
tive and Gram-negative bacteria and HGT is thought to be the main species being streptococci, and exam-
may be a “universal language” for mechanism in acquiring antibiotic ples include mutacin produced by
inter-species and inter-kingdom com- resistance genes (ARGs), which are S. mutans (Merritt & Qi 2012), san-
munication in dental biofilms, and richly present in the oral cavity guicin by S. sanguinis and salivaricin
the efficiency of signalling might be (Sukumar et al. 2016). by S. salivarius (Jakubovics et al.
enhanced by co-adhesion. Biofilm As described earlier, signalling 2014). Two types of mutacin have
formation with two co-adhering spe- molecules such as CSP markedly been detected; lantibiotics, which
cies – S. oralis and Actinomyces increase the ability of recipient cells have a broad spectrum of activity,
naeslundii – was inhibited when an to take up DNA (Senadheera & and the more common non-lantibio-
AI-2 knockout of S. oralis was used Cvitkovitch 2008). Extracellular tics, which have a narrower antimi-
instead of the wild type (Rickard DNA (eDNA) is a component of the crobial range (Merritt & Qi 2012).
et al. 2006), while AI-2 produced by biofilm matrix and plays a critical Lactobacilli also produce bacteri-
Aggregatibacter actinomycetemcomi- role in adhesion and in possible ocins and are being evaluated as
tans inhibited hyphae formation and nutrient storage and as a potential potential oral probiotics largely due
biofilm formation by C. albicans source of phosphate and other ions to their antimicrobial properties; for
(Bachtiar et al. 2014). AI-2 produced (Jakubovics & Burgess 2015). eDNA example, reuterin from Lactobacillus
by F. nucleatum had a differential release has been demonstrated in reuteri was active against selected
effect on biofilm formation when dual species experiments with periodontal and cariogenic bacteria
cultured with two different species of S. mutans and S. gordonii through (Kang et al. 2011).
oral streptococci; biofilm formation S. mutans competence-induced Bacterial “warfare” implies that
was enhanced with S. gordonii but bacteriocin production (Kreth et al. one of the interacting partners bene-
reduced with S. oralis (Jang et al. 2005); Gram-negative bacteria also fits at the expense of the other. This
2013). Some of these responses are release eDNA, including Veillonella has been shown with two taxa occu-
spp. (Hannan et al. 2010), pying the same niche – S. gordonii
© 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
S16 Marsh and Zaura

and S. mutans, where S. gordonii


had a competitive advantage over
S. mutans when using amino sugars
from salivary glycoproteins as an
energy source: S. gordonii released
hydrogen peroxide that inhibited
transcription of S. mutans genes
responsible for the metabolism of
these compounds (Zeng et al. 2016).
Indeed, hydrogen peroxide is one of
the most studied agents produced in
dental biofilms, but its impact on the
oral microbiota is complex and diffi-
cult to predict. Under aerobic condi-
tions (as could occur during early
stages of biofilm formation), Strepto-
coccus sanguinis produces high con-
centrations of hydrogen peroxide
that are capable of inhibiting a range
of Gram-positive species (Holmberg
Fig. 2. Model for Streptococcus gordonii and Aggregatibacter actinomycetemcomitans
& Hallander 1972, 1973, Kreth et al. interactions: hydrogen peroxide production by S. gordonii (Sg) supports lactate con-
2016); much lower concentrations sumption by A. actinomycetemcomitans (Aa) (figure S8 from Stacy et al. 2014).
are generated during anaerobic A. actinomycetemcomitans expresses H2O2-detoxifying enzyme catalase (KatA), which
growth. Streptococcus mutans is sus- also protects S. gordonii from self-inflicted oxidative stress. Dispersin B (DspB) is an
ceptible to hydrogen peroxide, but enzyme that promotes dispersal of A. actinomycetemcomitans biofilms and results in
strains that produce mutacin are increased distance between the A. actinomycetemcomitans and the H2O2-producing
able to inhibit other streptococci S. gordonii. The three zones (Peroxide killing zone, Synergy zone and Carbon starva-
(Ryan & Kleinberg 1995, Ashby tion zone) correspond to different concentrations in oxygen, hydrogen peroxide and
et al. 2009). Hydrogen peroxide pro- lactate in the biofilm, as indicated with the respective triangles.
duction has been proposed as a
major mechanism for controlling the elevated H2O2 by induction of Dis- microbial community structure in
levels of putative periodontopathic persin B – an enzyme that promotes health that was absent during peri-
bacteria in dental plaque (Hillman dispersal of A. actinomycetemcomi- odontal disease (Wang et al. 2016).
et al. 1985, Hillman & Shivers 1988). tans biofilms, resulting in increased Antagonism will also be a mecha-
However, other bacteria in the physical distance between the nism whereby exogenous species are
supragingival biofilms (e.g. Neisseria, A. actinomycetemcomitans and the prevented from colonizing the oral
Haemophilus and Actinomyces spe- H2O2-producing S. gordonii. On the cavity (bacterial interference or colo-
cies) are also able to degrade hydro- other hand, S. gordonii, which does nization resistance). Oral strepto-
gen peroxide, and little free peroxide not make its own catalase, is cross- cocci have been shown to interfere
can be detected in plaque (Ryan & protected by A. actinomycetemcomi- with colonization by Pseudomonas
Kleinberg 1995). Thus, there may be tans from self-inflicted oxidative aeruginosa through nitrite-mediated
varying concentrations of hydrogen stress. interference (Scoffield & Wu 2015,
peroxide in different regions of the A highly diverse oral bacterio- 2016), while a sophisticated coloniza-
biofilm, and the balance between phage gene pool has been discovered tion resistance structure has been
symbiosis and dysbiosis may depend through a metagenomics approach described in an in vitro murine oral
on the complex interplay between (Pride et al. 2012, Naidu et al. 2014, microbial community with the “Sen-
multiple antagonistic microbial inter- Dalmasso et al. 2015, Edlund et al. sor” (Streptococcus saprophyticus)
actions. 2015a). Phages are bacterial viruses sensing the intruding non-oral
Counter-intuitively, antagonistic that may lyse competing cells. The Escherichia coli strain and producing
interactions might also be beneficial production of antagonistic factors diffusible signals to the “Mediator”
to both partners involved and might will not necessarily lead to the com- (Streptococcus infantis) that de-
even stimulate the fitness of the plete exclusion of sensitive species as represses the capacity of the “Killer”
microbial community (Stacy et al. the presence of distinct microhabi- (Streptococcus sanguinis) to produce
2014). In the presence of oxygen, tats within a biofilm such as plaque hydrogen peroxide, resulting in inhi-
A. actinomycetemcomitans that cross- enable bacteria to survive under con- bition of the invading E. coli (He
feeds with lactate produced by ditions that would be incompatible et al. 2014).
S. gordonii has to survive high con- to them in a homogeneous environ-
centrations of hydrogen peroxide ment. Noteworthy, although para-
Ecological drivers towards dysbiosis
released by S. gordonii (Fig. 2). To sitic by their nature, phages might
and disease
ameliorate oxidative stress, A. acti- have a beneficial role in the oral
nomycetemcomitans not only ecosystem: a recent comparison of When the oral environment changes,
expresses catalase (H2O2-detoxifying the bacteria-phage network revealed the ecology of the ecosystem is
enzyme), but also responds to that phages supported a complex affected. This has an impact on the
© 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Oral microbial interactions S17

outcome of the interactions among dysbiosis results not only in reduced as P. gingivalis (Olczak et al. 2005).
the microorganisms in the biofilms, taxonomic diversity, but also in a The change in local environmental
which will affect the proportions of changed metaproteome, as recently conditions associated with inflamma-
the members of the community, and shown in microcosms where proteins tion will alter the competitiveness
can increase the risk of disease (dys- involved in acid tolerance and acid and outcome of multiple interactions
biosis). Two scenarios will be dis- production dominated the dysbiotic among the microbes that make up
sected below: one leading towards a biofilms (Rudney et al. 2015). the subgingival microbiota, leading
cariogenic and the other towards a A counter mechanism against to substantial changes in the micro-
periodontopathogenic ecosystem. acidification of the ecosystem is bial composition of the biofilm.
Dental caries is associated with alkali production by the members of Although there is agreement that
an increased frequency of dietary the community, mainly through there are major changes in the pro-
sugar intake. These sugars are ammonia production from arginine portions of individual species in bio-
metabolized rapidly to acid (mainly and urea (Burne & Marquis 2000, films from inflamed sites (e.g. see
lactic acid), and a low pH is gener- Shu et al. 2003, Liu et al. 2012, reviews by Perez-Chaparro et al.
ated within the biofilm. Lactate can Huang et al. 2015, Takahashi 2015). 2014, Diaz et al. 2016), there are
be utilized by Veillonella spp., and Recently, by applying a metatran- conflicting reports on whether the
other species, for example Neisseria scriptomics and metabolomics diversity of the resultant microbial
(Hoshino & Araya 1980), Hae- approach, a much higher diversity in communities is altered. The diversity
mophilus (Traudt & Kleinberg 1996), alkali-generating pathways within may increase in gingivitis (Kistler
Aggregatibacter (Brown & Whiteley complex oral biofilms has been dis- et al. 2013, Schincaglia et al. 2017),
2007), Porphyromonas (Lewis et al. covered, including glutamate dehy- but the evidence for chronic peri-
2009) and Actinomyces (Takahashi & drogenase, threonine and serine odontitis is more contentious (Abus-
Yamada 1996), and converted to deaminase, and upregulation in leme et al. 2013, Hong et al. 2015,
weaker acids. Fewer carious lesions membrane proteins involved in Kirst et al. 2015, Park et al. 2015).
and less lactate in plaque were mea- ammonia gas conduction besides the The inflammatory response can
sured in rats inoculated with S. mu- urease activity and arginine deimi- influence the subgingival microbiota
tans and Veillonella alcalescens than nase system (Edlund et al. 2015b). in two ways: (i) via the impact of the
in animals infected with S. mutans Additionally, this study revealed that host defences and (ii) by the resul-
alone (van der Hoeven et al. 1978). Veillonella species are well adapted tant changes to the environment.
Higher proportions of Veillonella towards acid stress by upregulating The innate defences will inhibit sus-
spp. have been detected in samples various pathways that contributed to ceptible species, but a number of
from caries lesions when compared pH recovery. periodontal pathogens, such as
to plaque from healthy enamel Thus, unlike health, dental caries P. gingivalis, can subvert the host
(Gross et al. 2012), perhaps because is associated with a shift in the com- response, for example, by degrading
of the increased glycolytic activity position of the biofilm to a commu- complement, interfering with neu-
and higher levels of lactate at these nity that is dominated by a strongly trophil function and blocking phago-
sites. Symbiosis between Veillonella saccharolytic and acid-tolerant cytosis (for reviews, see Slaney &
and S. mutans has been demon- microbiota leading to a loss of diver- Curtis 2008, Hajishengallis &
strated in mixed cultures: when Veil- sity, and a reduction in levels and Lamont 2014, Mysak et al. 2014).
lonella parvula was added to the pair activity of beneficial bacteria (Gross Thus, sensitive species will be elimi-
of antagonists (S. mutans and et al. 2012, Jiang et al. 2011, Li nated (although some may survive
S. gordonii), it mitigated the inhibi- et al. 2007, Peterson et al. 2013), due to cross-protection from neigh-
tory effects of S. gordonii on sugar although the diversity may increase bouring organisms), but those that
metabolism and growth of S. mutans when the lesion penetrates dentine, can tolerate the inflammatory
(Liu et al. 2011). perhaps reflecting important environ- response will flourish. It has been
The frequent conditions of low mental changes (Sim on-Soro et al. argued that the microbial consortia
pH in biofilms associated with caries 2014). that are associated with periodontitis
are inhibitory to the growth of many In contrast, the accumulation of are “inflammo-philic” in that they
of the bacteria associated with microbial biomass around the gingi- have adapted to not only endure
enamel health, resulting in decreased val margin induces an inflammatory inflammation but also to exploit the
microbial diversity (Gross et al. response. This results in an increased altered environmental conditions
2012, Jiang et al. 2011, Li et al. flow of GCF, which delivers not (Hajishengallis 2014), such as small
2007, Peterson et al. 2013). Repeated only components of the host rises in pH and temperature (Eggert
conditions of low pH alter the com- defences (e.g. immunoglobulins, et al. 1991, Fedi & Killoy 1992, Haf-
petitiveness of members of the bio- complement, neutrophils, cytokines) fajee et al. 1992, Nyako et al. 2005).
film community and select for (Ebersole 2003), but, inadvertently, Such small changes to the local envi-
increased proportions of acidogenic host molecules that can act as sub- ronment can alter gene expression
and acid-tolerating bacteria includ- strates for proteolytic bacteria. Some and increase the competitiveness of
ing mutans streptococci, lactobacilli of these host molecules also contain species such as P. gingivalis within
(Bradshaw et al. 1989), low-pH non- haemin (e.g. haptoglobin, haemo- microbial communities (Marsh et al.
S. mutans streptococci and bifi- pexin, haemoglobin), which is an 1993). However, a more substantial
dobacteria (Marsh 1994, Takahashi essential cofactor for the growth of change to the inflamed pocket is the
& Nyvad 2008). Sucrose-induced potential periodontopathogens such altered nutrient status as a result of
© 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
S18 Marsh and Zaura

the increased flow of GCF. To study have demonstrated the importance should be susceptible to the action
the impact of this, laboratory studies of close physical associations to bio- of the drug (Walker et al. 1987, van
have been performed using serum as film formation by interacting species Winkelhoff et al. 1997, Rams et al.
a surrogate for GCF, and complex of Gram-negative anaerobic bacteria 2013).
nutritional inter-relationships among (Sharma et al. 2005, Okuda et al. Attempts have also been made to
subgingivally derived microbial con- 2012). exploit antagonistic interactions to
sortia have been observed (ter Steeg Periodontal diseases may be an resolve both periodontal disease and
et al. 1987, ter Steeg & van der Hoe- example of “pathogenic synergism” caries. For periodontal therapy,
ven 1989). When biofilms from (van Steenbergen et al. 1984), in either bacterial interference has been
patients with chronic periodontitis which disease is a consequence of the applied by deliberately implanting
were inoculated into pre-reduced (i.e. combined activity of an interacting beneficial oral bacteria into a treated
anaerobic) heat-inactivated human consortium in which each member is pocket (van Essche et al. 2013,
serum, the microbial composition of only weakly virulent. Different spe- Teughels et al. 2013) or using preda-
the consortia changed over time and cies would undertake a distinct role tory protozoa, such as Bdellovibrio
these changes correlated with distinct or function in order for the consor- species (Dashiff & Kadouri 2011, van
stages in glycoprotein breakdown tium to persist and cause disease. This Essche et al. 2011, Loozen et al.
involving bacteria with different is consistent with the recent concept 2015) or bacteriophage (reviewed by
metabolic capabilities. Initially, car- of low abundance species (“keystone Allaker & Douglas 2009), while for
bohydrate side chains were removed pathogens”) having a disproportion- caries prevention, different
by organisms with complementary ate effect of the virulence of the whole approaches (e.g. lozenges, milk,
glycosidase activities; this was fol- community (Hajishengallis et al. yoghurt) with probiotic bacteria that
lowed by the hydrolysis of the pro- 2011, Hajishengallis & Lamont 2012). are antagonistic against S. mutans
tein core by obligately anaerobic Gene transfer can occur within these have been tried (Cagetti et al. 2013).
bacteria leading to extensive amino communities; this can include not A recent systematic review on the use
acid fermentation. Significantly, indi- only mobile elements that code for of probiotics in managing oral dis-
vidual species grew only poorly in drug resistance but also larger eases concluded that there is sufficient
pure culture on serum (ter Steeg & stretches of DNA that effect the viru- evidence for supporting the use of
van der Hoeven 1989). lence of recipient cells, for example, probiotics in the case of gingivitis and
Numerous nutritional inter- P. gingivalis possesses a “pathogenic- periodontitis but not for caries (Gru-
dependencies and physical interac- ity island” (Curtis et al. 1999). ner et al. 2016), although this is an
tions will develop among the species Evidence for the role of the entire area in which more research is
coping with the array of novel host community and not just a few patho- required.
factors produced during the inflam- gens in dysbiosis has recently been
matory response. For example, a delivered by metatranscriptome anal-
Conclusions
complex but symbiotic metabolic ysis of dental biofilms from sites with
relationship has been demonstrated active periodontal disease (Yost et al. Microbial communities, such as
in laboratory studies of P. gingivalis 2015): various streptococci, Veil- those found in dental biofilms, dis-
and Treponema denticola (Grenier lonella parvula and Pseudomonas fluo- play “emergent properties”; that is,
1992, Tan et al. 2014). Early studies rescens, were highly active in their properties are more than the
demonstrated that isobutyric acid transcribing putative virulence factors sum of the component species, and
produced by P. gingivalis stimulated besides periodontal pathogens such their characteristics cannot be
the growth of T. denticola, while suc- as Tannerella forsythia and P. gingi- inferred from studies of individual
cinic acid generated by T. denticola valis. The genes that were over-repre- organisms (Diaz et al. 2014). The
enhanced the growth of P. gingivalis sented at these sites were related to microbiota is structurally and func-
(Grenier 1992). More recent studies cell motility, lipid A and peptidogly- tionally organized, and it has been
have shown that the biomass is can biosynthesis, and transport of argued that such microbial commu-
higher when both species are grown iron, potassium and amino acids. nities could be considered as primi-
in co-culture, and glycine produced Microbial interactions in such tive multicellular organisms
by P. gingivalis is utilized by the complex consortia could influence (Caldwell et al. 1997, Ereshefsky &
spirochaete (Tan et al. 2014). Both treatment outcomes. Although not Pedroso 2015). In health, numerous
species respond to the presence of advocated for routine use in peri- interactions contribute to stability
the other as seen by changes in glo- odontal disease, antibiotics are fre- and resilience of the ecosystem
bal gene expression in both species. quently used as adjunctive treatment against environmental perturbations
Similarly, the growth of certain spe- to mechanical debridement in cases (Alexander 1971, Marsh 1989).
cies that have been previously with severe or recurrent disease (Jep- If certain key environmental pres-
described as being “unculturable” sen & Jepsen 2016). However, care sures exceed thresholds that vary
(e.g. Fretibacterium fastidiosum, Pre- needs to be taken as, apart from the from patient to patient, then the
votella HOT-376, Tannerella HOT- existence and inter-species transfer of competitiveness of certain bacteria is
286) has been shown recently to be resistance genes within microbial altered and dysbiosis can occur,
due to their dependence on sidero- communities, b-lactamase producing leading to caries or periodontal dis-
phores and to the close physical bacteria are commonly present in eases. In caries and periodontal dis-
proximity of “helper” strains (Var- subgingival biofilms and they could eases, changes in the nutrient status
toukian et al. 2016a,b). Other studies protect neighbouring organisms that at the site due to increases in
© 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Oral microbial interactions S19

fermentable carbohydrates (and the Amsterdam for her advice on the evolution and spread of a major class of
mosaic pbp2x in Streptococcus pneumoniae, S.
resultant acidic conditions) and host search strategy.
mitis and S. oralis. International Journal of
proteins (including haemin-contain- Medical Microbiology 297, 503–512.
ing molecules), respectively, disrupt Curtis, M. A., Hanley, S. A. & Aduse-Opoku, J.
the microbial interactions that con- References (1999) The rag locus of Porphyromonas gingi-
trol the balance of the microbial valis: a novel pathogenicity island. Journal of
Abusleme, L., Dupuy, A. K., Dutzan, N., Silva,
Periodontal Research 34, 400–405.
communities in health. Effective pre- N., Burleson, J. A., Strausbaugh, L. D., Gamo-
Dalmasso, M., de Haas, E., Neve, H., Strain, R.,
vention of dental disease will require nal, J. & Diaz, P. I. (2013) The subgingival
Cousin, F. J., Stockdale, S. R., Ross, R. P. &
microbiome in health and periodontitis and its
interference with these factors that relationship with community biomass and
Hill, C. (2015) Isolation of a novel phage with
drive dysbiosis (Marsh 2003), and a activity against Streptococcus mutans biofilms.
inflammation. ISME Journal 7, 1016–1025.
PLoS One 10, e0138651.
greater understanding of microbial Alexander, M. (1971). Microbial Ecology, pp.
Dashiff, A. & Kadouri, D. E. (2011) Predation of
interactions could lead to strategies 207–223. London: Wiley.
oral pathogens by Bdellovibrio bacteriovorus
Ali Mohammed, M. M., Nerland, A. H., Al-Har-
to actively promote oral health. oni, M. & Bakken, V. (2013) Characterization
109J. Molecular Oral Microbiology 26, 19–34.
The current literature search led Diaz, P. I. (2012) Microbial diversity and interac-
of extracellular polymeric matrix, and treat-
tions in subgingival biofilm communities. Fron-
us to the following conclusions: (i) ment of Fusobacterium nucleatum and Porphy-
tiers of Oral Biology 15, 17–40.
oral microbial interactions belong to romonas gingivalis biofilms with DNase I and
Diaz, P. I., Hoare, A. & Hong, B. Y. (2016)
a highly studied and diverse topic, proteinase K. Journal of Oral Microbiology 5:
Subgingival microbiome shifts and community
20015. doi:10.3402/jom.v5i0.20015.
which was too broad for a system- Allaker, R. P. & Douglas, C. W. I. (2009) Novel
dynamics in periodontal diseases. Journal of the
California Dental Association 44: 421–435.
atic review; (ii) most oral microbial anti-microbial therapies for dental plaque-
Diaz, P. I., Strausbaugh, L. D. & Dongari-Bagt-
interactions have been investigated related diseases. International Journal of
zoglou, A. (2014) Fungal-bacterial interactions
in laboratory systems, and occasion- Antimicrobial Agents 33, 8–13.
and their relevance to oral health: linking the
Ashby, M. T., Kreth, J., Soundarajan, M. &
ally animal models, and therefore Sivuilu, L. S. (2009) Influence of a model
clinic and the bench. Frontiers in Cellular and
some caution should be exercised Infection Microbiology 4: 101. doi:10.3389/
human defensive peroxidase system on oral
fcimb.2014.00101.
when extrapolating these findings to streptococcal antagonism. Microbiology 155,
Diaz, P. I., Xie, Z., Sobue, T., Thompson, A., Biyi-
events in humans; (iii) the majority 3691–3700.
koglu, B., Ricker, A., Ikonomou, L. & Dongari-
Bachtiar, E. W., Bachtiar, B. M., Jarosz, L. M.,
of the interactions studied involve Amir, L. R., Sunarto, H., Ganin, H., Meijler,
Bagtzoglou, A. (2012) Synergistic interaction
bacteria only, while other segments between Candida albicans and commensal oral
M. M. & Krom, B. P. (2014) Ai-2 of Aggregat-
streptococci in a novel in vitro mucosal model.
of the oral microbiota (fungi, ibacter actinomycetemcomitans inhibits Candida
Infection and Immunity 80, 620–632.
Archaea, viruses, protozoa) are albicans biofilm formation. Frontiers in Cellular
Diaz, P. I., Zilm, P. S. & Rogers, A. H. (2002)
and Infection Microbiology 4, 94.
understudied; (iv) current technologi- Bergeron, L. J. & Burne, R. A. (2001) Roles of
Fusobacterium nucleatum supports the growth
cal advances (e.g. metagenomics, of Porphyromonas gingivalis in oxygenated and
fructosyltransferase and levanase-sucrase of
carbon-dioxide-depleted environments. Microbi-
metatranscriptomics, metapro- Actinomyces naeslundii in fructan and sucrose
ology 148, 467–472.
teomics, metabolomics, spectral metabolism. Infection and Immunity 69, 5395–
Dige, I., Gronkjar, L. & Nyvad, B. (2014) Molec-
imaging fluorescence in situ 5402.
ular studies of the structural ecology of natural
Bowler, L. D., Zhang, Q. Y., Riou, J. Y. &
hybridization) enable the study of Spratt, B. G. (1994) Interspecies recombination
occlusal caries. Caries Research 48, 451–460.
Dige, I., Nilsson, H., Kilian, M. & Nyvad, B.
more complex community level inter- between the pena genes of Neisseria meningi-
(2007) In situ identification of streptococci and
actions, including those among tidis and commensal Neisseria species during
other bacteria in initial dental biofilm by confo-
members of the microbiota from dif- the emergence of penicillin resistance in N.
cal laser scanning microscopy and fluorescence
meningitidis: natural events and laboratory sim-
ferent kingdoms (Diaz et al. 2014) ulation. Journal of Bacteriology 176, 333–337.
in situ hybridization. European Journal of Oral
rather than just the conventional Sciences 115, 459–467.
Bradshaw, D. J., Homer, K. A., Marsh, P. D. &
Ebersole, J. L. (2003) Humoral immune responses
dual species studies; (v) both syner- Beighton, D. (1994) Metabolic cooperation in
in gingival crevice fluid: local and systemic
gistic and antagonistic interactions oral microbial communities during growth on
implications. Periodontology 2000 31, 135–166.
mucin. Microbiology 140, 3407–3412.
contribute to the ecological stability Bradshaw, D. J., McKee, A. S. & Marsh, P. D.
Edlund, A., Santiago-Rodriguez, T. M., Boehm,
of the microbial community that T. K. & Pride, D. T. (2015a) Bacteriophage
(1989) Effects of carbohydrate pulses and pH
and their potential roles in the human oral cav-
characterizes oral health; and (vi) on population shifts within oral microbial com-
ity. Journal of Oral Microbiology 7, 27423.
more attention needs to be focussed munities in vitro. Journal of Dental Research
Edlund, A., Yang, Y., Yooseph, S., Hall, A. P.,
68, 1298–1302.
on what microorganisms are doing Brown, S. A. & Whiteley, M. (2007) A novel
Nguyen, D. D., Dorrestein, P. C., Nelson, K.
within these microbial communities E., He, X., Lux, R., Shi, W. & McLean, J. S.
exclusion mechanism for carbon resource parti-
(2015b) Meta-omics uncover temporal regula-
(Takahashi 2015), rather than just tioning in Aggregatibacter actinomycetemcomi-
tion of pathways across oral microbiome gen-
cataloguing which ones are present. tans. Journal of Bacteriology 189, 6407–6414.
era during in vitro sugar metabolism. ISME
Burne, R. A. & Marquis, R. E. (2000) Alkali pro-
The oral microbiome in health and Journal 9, 2605–2619.
duction by oral bacteria and protection against
disease might be better described by dental caries. FEMS Microbiology Letters 193,
Eggert, F. M., Drewell, L., Bigelow, J. A., Speck,
J. E. & Goldner, M. (1991) The pH of gingival
a series of functions and interac- 1–6.
crevices and periodontal pockets in children,
tions, rather than as a list of individ- Cagetti, M. G., Mastroberardino, S., Milia, E.,
teenagers and adults. Archives of Oral Biology
ual organisms, as these functions Cocco, F., Lingstrom, P. & Campus, G. (2013)
36, 233–238.
The use of probiotic strains in caries preven-
might not be provided by the same tion: a systematic review. Nutrients 5, 2530–
Ereshefsky, M. & Pedroso, M. (2015) Rethinking
microbes in different people (Lloyd- evolutionary individuality. Proceedings of the
2550.
National Academy of Sciences of the United
Price et al. 2016). Caldwell, D. E., Wolfaardt, G. M., Korber, D. R.
States of America 112, 10126–10132.
& Lawrence, J. R. (1997). Do bacterial commu-
van Essche, M., Loozen, G., Godts, C., Boon, N.,
nities transcend darwinism? In: Jones, J. G.
Pauwels, M., Quirynen, M. & Teughels, W.
Acknowledgement (ed.). Advances in Microbial Ecology, pp. 105–
(2013) Bacterial antagonism against periodon-
191, Boston, MA: Springer US.
topathogens. Journal of Periodontology 84,
We are thankful to medical informa- Chi, F., Nolte, O., Bergmann, C., Ip, M. & Hak-
801–811.
tion specialist Ilse Jansma at VUmc enbeck, R. (2007) Crossing the barrier:

© 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
S20 Marsh and Zaura

van Essche, M., Quirynen, M., Sliepen, I., Loo- biofilm development. Journal of Dental Kistler, J. O., Booth, V., Bradshaw, D. J. &
zen, G., Boon, N., van Eldere, J. & Teughels, Research 88, 982–990. Wade, W. G. (2013) Bacterial community
W. (2011) Killing of anaerobic pathogens by Holmberg, K. & Hallander, H. O. (1972) Interfer- development in experimental gingivitis. PLoS
predatory bacteria. Molecular Oral Microbiol- ence between gram-positive microorganisms in One 8, e71227.
ogy 26, 52–61. dental plaque. Journal of Dental Research 51, Kolenbrander, P. E. (2011) Multispecies commu-
Fedi, P. F. & Killoy, W. J. (1992) Temperature 588–595. nities: interspecies interactions influence growth
differences at periodontal sites in health and Holmberg, K. & Hallander, H. O. (1973) Produc- on saliva as sole nutritional source. Interna-
disease. Journal of Periodontology 63, 24–27. tion of bactericidal concentrations of hydrogen tional Journal of Oral Science 3, 49–54.
Grenier, D. (1992) Nutritional interactions peroxide by Streptococcus sanguis. Archives of Koo, H., Falsetta, M. L. & Klein, M. I. (2013)
between two suspected periodontopathogens, Oral Biology 18, 423–434. The exopolysaccharide matrix: a virulence
Treponema denticola and Porphyromonas gingi- Homer, K. A. & Beighton, D. (1992) Synergistic determinant of cariogenic biofilm. Journal of
valis. Infection and Immunity 60, 5298–5301. degradation of bovine serum albumin by mutans Dental Research 92, 1065–1073.
Gross, E. L., Beall, C. J., Kutsch, S. R., Fire- streptococci and other dental plaque bacteria. Kreth, J., Giacaman, R. A., Raghavan, R. &
stone, N. D., Leys, E. J. & Griffen, A. L. FEMS Microbiology Letters 69, 259–262. Merritt, J. (2016) The road less traveled –
(2012) Beyond Streptococcus mutans: dental Hong, B.-Y., Furtado Araujo, M. V., Straus- defining molecular commensalism with Strepto-
caries onset linked to multiple species by 16S baugh, L. D., Terzi, E., Ioannidou, E. & Diaz, coccus sanguinis. Molecular Oral Microbiology
rRNA community analysis. PLoS One 7, P. I. (2015) Microbiome profiles in periodonti- doi: 10.1111/omi.12170.
e47722. tis in relation to host and disease characteris- Kreth, J., Merritt, J., Shi, W. & Qi, F. (2005) Co-
Gruner, D., Paris, S. & Schwendicke, F. (2016) tics. PLoS One 10, e0127077. ordinated bacteriocin production and compe-
Probiotics for managing caries and periodonti- Hoshino, E. & Araya, A. (1980) Lactate degrada- tence development: a possible mechanism for
tis: systematic review and meta-analysis. Jour- tion by polysaccharide-producing Neisseria iso- taking up DNA from neighbouring species.
nal of Dentistry 48, 16–25. lated from human dental plaque. Archives of Molecular Microbiology 57, 392–404.
Guo, L., He, X. & Shi, W. (2014) Intercellular Oral Biology 25, 211–212. Lewis, J. P., Iyer, D. & Anaya-Bergman, C.
communications in multispecies oral microbial Huang, R., Li, M. & Gregory, R. L. (2011) Bacte- (2009) Adaptation of Porphyromonas gingivalis
communities. Frontiers in Microbiology 5, 328. rial interactions in dental biofilm. Virulence 2, to microaerophilic conditions involves
Haffajee, A. D., Socransky, S. S. & Goodson, J. 435–444. increased consumption of formate and reduced
M. (1992) Subgingival temperature (i). Relation Huang, X., Schulte, R. M., Burne, R. A. & Nasci- utilization of lactate. Microbiology 155, 3758–
to baseline clinical parameters. Journal of Clini- mento, M. M. (2015) Characterization of the 3774.
cal Periodontology 19, 401–408. arginolytic microflora provides insights into ph Li, Y., Ge, Y., Saxena, D. & Caufield, P. W.
Hajishengallis, G. (2014) Immunomicrobial homeostasis in human oral biofilms. Caries (2007) Genetic profiling of the oral microbiota
pathogenesis of periodontitis: keystones, patho- Research 49, 165–176. associated with severe early-childhood caries.
bionts, and host response. Trends in Immunol- Jack, A. A., Daniels, D. E., Jepson, M. A., Vick- Journal of Clinical Microbiology 45, 81–87.
ogy 35, 3–11. erman, M. M., Lamont, R. J., Jenkinson, H. Liu, J., Wu, C., Huang, I.-H., Merritt, J. & Qi, F.
Hajishengallis, G. & Lamont, R. J. (2012) Beyond F. & Nobbs, A. H. (2015) Streptococcus gor- (2011) Differential response of Streptococcus
the red complex and into more complexity: the donii comCDE (competence) operon modulates mutans towards friend and foe in mixed-species
polymicrobial synergy and dysbiosis (PSD) biofilm formation with Candida albicans. cultures. Microbiology 157, 2433–2444.
model of periodontal disease etiology. Molecu- Microbiology 161, 411–421. Liu, Y.-L., Nascimento, M. & Burne, R. A.
lar Oral Microbiology 27, 409–419. Jakubovics, N. S. (2015a) Intermicrobial interac- (2012) Progress toward understanding the con-
Hajishengallis, G. & Lamont, R. J. (2014) Break- tions as a driver for community composition tribution of alkali generation in dental biofilms
ing bad: manipulation of the host response by and stratification of oral biofilms. Journal of to inhibition of dental caries. International
Porphyromonas gingivalis. European Journal of Molecular Biology 427, 3662–3675. Journal of Oral Science 4, 135–140.
Immunology 44, 328–338. Jakubovics, N. S. (2015b) Saliva as the sole nutri- Lloyd-Price, J., Abu-Ali, G. & Huttenhower, C.
Hajishengallis, G., Liang, S., Payne, M. A., tional source in the development of multi- (2016) The healthy human microbiome. Gen-
Hashim, A., Jotwani, R., Eskan, M. A., McIn- species communities in dental plaque. ome Medicine 8, 51.
tosh, M. L., Alsam, A., Kirkwood, K. L., Microbiology Spectrum 3, doi: 10.1128/micro Loozen, G., Boon, N., Pauwels, M., Slomka, V.,
Lambris, J. D., Darveau, R. P. & Curtis, M. biolspec.MBP-0013-2014. Rodrigues Herrero, E., Quirynen, M. & Teugh-
A. (2011) Low-abundance biofilm species Jakubovics, N. S. & Burgess, J. G. (2015) Extra- els, W. (2015) Effect of Bdellovibrio bacteri-
orchestrates inflammatory periodontal disease cellular DNA in oral microbial biofilms. ovorus HD100 on multispecies oral
through the commensal microbiota and com- Microbes and Infection 17, 531–537. communities. Anaerobe 35, 45–53.
plement. Cell Host & Microbe 10, 497–506. Jakubovics, N. S., Yassin, S. A. & Rickard, A. H. Mark Welch, J. L., Rossetti, B. J., Rieken, C. W.,
Hannan, S., Ready, D., Jasni, A. S., Rogers, M., (2014) Community interactions of oral strepto- Dewhirst, F. E. & Borisy, G. G. (2016) Bio-
Pratten, J. & Roberts, A. P. (2010) Transfer of cocci. Advances in Applied Microbiology 87, 43– geography of a human oral microbiome at the
antibiotic resistance by transformation with 110. micron scale. Proceedings of the National Acad-
eDNA within oral biofilms. FEMS Immunology Jang, Y.-J., Sim, J., Jun, H.-K. & Choi, B.-K. emy of Sciences of the United States of America
and Medical Microbiology 59, 345–349. (2013) Differential effect of autoinducer 2 of 113, E791–E800.
He, X., McLean, J. S., Guo, L., Lux, R. & Shi, Fusobacterium nucleatum on oral streptococci. Marsh, P. (1989) Host defenses and microbial
W. (2014) The social structure of microbial Archives of Oral Biology 58, 1594–1602. homeostasis: role of microbial interactions.
community involved in colonization resistance. Jepsen, K. & Jepsen, S. (2016) Antibiotics/antimi- Journal of Dental Research 68, 1567–1575.
ISME Journal 8, 564–574. crobials: systemic and local administration in Marsh, P. D. (1994) Microbial ecology of dental
Hillman, J. D. & Shivers, M. (1988) Interaction the therapy of mild to moderately advanced plaque and its significance in health and dis-
between wild-type, mutant and revertant forms periodontitis. Periodontology 2000 71, 82–112. ease. Advances in Dental Research 8, 263–271.
of the bacterium Streptococcus sanguis and the Jiang, W., Jiang, Y., Li, C. & Liang, J. (2011) Marsh, P. D. (2003) Are dental diseases examples
bacterium Actinobacillus actinomycetemcomi- Investigation of supragingival plaque of ecological catastrophes? Microbiology 149,
tans in vitro and in the gnotobiotic rat. Archives microbiota in different caries status of chinese 279–294.
of Oral Biology 33, 395–401. preschool children by denaturing gradient gel Marsh, P. D., McKee, A. S. & Mothershaw, A.
Hillman, J. D., Socransky, S. S. & Shivers, M. electrophoresis. Microbial Ecology 61, 342–352. S. (1993) Continuous culture studies of Porphy-
(1985) The relationships between streptococcal Kang, M.-S., Oh, J.-S., Lee, H.-C., Lim, H.-S., romonas gingivalis. In: Shah, H. N., Mayrand,
species and periodontopathic bacteria in human Lee, S.-W., Yang, K.-H., Choi, N.-K. & Kim, D. & Genco, R. J. (eds). Biology of the Species
dental plaque. Archives of Oral Biology 30, S.-M. (2011) Inhibitory effect of Lactobacillus Porphyromonas gingivalis, pp. 105–123, Boca
791–795. reuteri on periodontopathic and cariogenic bac- Raton, FL: CRC Press.
van der Hoeven, J. S., Toorop, A. I. & Mikx, R. teria. Journal of Microbiology 49, 193–199. Mazumdar, V., Amar, S. & Segre, D. (2013)
H. (1978) Symbiotic relationship of Veillonella Kirst, M. E., Li, E. C., Alfant, B., Chi, Y.-Y., Metabolic proximity in the order of coloniza-
alcalescens and Streptococcus mutans in dental Walker, C., Magnusson, I. & Wang, G. P. tion of a microbial community. PLoS One 8,
plaque in gnotobiotic rats. Caries Research 12, (2015) Dysbiosis and alterations in predicted e77617.
142–147. functions of the subgingival microbiome in Merritt, J. & Qi, F. (2012) The mutacins of Strep-
Hojo, K., Nagaoka, S., Ohshima, T. & Maeda, chronic periodontitis. Applied and Environment tococcus mutans: regulation and ecology.
N. (2009) Bacterial interactions in dental Microbiology 81, 783–793. Molecular Oral Microbiology 27, 57–69.

© 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
Oral microbial interactions S21

Moye, Z. D., Zeng, L. & Burne, R. A. (2014) Fuel- Rickard, A. H., Palmer, R. J., Blehert, D. S., ter Steeg, P. F., van der Hoeven, J. S., de Jong,
ing the caries process: carbohydrate metabolism Campagna, S. R., Semmelhack, M. F., Egland, M. H., van Munster, P. J. & Jansen, M. J.
and gene regulation by Streptococcus mutans. P. G., Bassler, B. L. & Kolenbrander, P. E. (1987) Enrichment of subgingival microflora on
Journal of Oral Microbiology 6, doi: 10.3402/ (2006) Autoinducer 2: a concentration-depen- human serum leading to accumulation of Bac-
jom.v6.24878. dent signal for mutualistic bacterial biofilm teroides species, peptostreptococci and
Mysak, J., Podzimek, S., Sommerova, P., Lyuya- growth. Molecular Microbiology 60, 1446–1456. Fusobacteria. Antonie van Leeuwenhoek 53,
Mi, Y., Bartova, J., Janatova, T., Prochazkova, Roberts, A. P. & Kreth, J. (2014) The impact 261–272.
J. & Duskova, J. (2014) Porphyromonas gingi- of horizontal gene transfer on the adaptive van Steenbergen, T. J., van Winkelhoff, A. J. &
valis: major periodontopathic pathogen over- ability of the human oral microbiome. Fron- de Graaff, J. (1984) Pathogenic synergy: mixed
view. Journal of Immunology Research 2014, tiers in Cellular and Infection Microbiology 4, infections in the oral cavity. Antonie van
476068. 124. Leeuwenhoek 50, 789–798.
Naidu, M., Robles-Sikisaka, R., Abeles, S. R., Roberts, F. A. & Darveau, R. P. (2015) Microbial Sukumar, S., Roberts, A. P., Martin, F. E. &
Boehm, T. K. & Pride, D. T. (2014) Character- protection and virulence in periodontal tissue Adler, C. J. (2016) Metagenomic insights into
ization of bacteriophage communities and as a function of polymicrobial communities: transferable antibiotic resistance in oral bacte-
CRISPR profiles from dental plaque. BMC symbiosis and dysbiosis. Periodontology 2000 ria. Journal of Dental Research 95, 969–976.
Microbiology 14, 175. 69, 18–27. Takahashi, N. (2015) Oral microbiome metabo-
Ng, H. M., Kin, L. X., Dashper, S. G., Slakeski, Rudney, J. D., Jagtap, P. D., Reilly, C. S., Chen, lism: from “who are they?” to “what are they
N., Butler, C. A. & Reynolds, E. C. (2016) R., Markowski, T. W., Higgins, L., Johnson, J. doing?”. Journal of Dental Research 94, 1628–
Bacterial interactions in pathogenic subgingival E. & Griffin, T. J. (2015) Protein relative abun- 1637.
plaque. Microbial Pathogenesis 94, 60–69. dance patterns associated with sucrose-induced Takahashi, N. & Nyvad, B. (2008) Caries ecology
Nobbs, A. H. & Jenkinson, H. F. (2015) Interk- dysbiosis are conserved across taxonomically revisited: microbial dynamics and the caries
ingdom networking within the oral micro- diverse oral microcosm biofilm models of den- process. Caries Research 42, 409–418.
biome. Microbes and Infection 17, 484–492. tal caries. Microbiome 3, 69. Takahashi, N. & Yamada, T. (1996) Catabolic
Nyako, E. A., Watson, C. J. & Preston, A. J. Ryan, C. S. & Kleinberg, I. (1995) Bacteria in pathway for aerobic degradation of lactate by
(2005) Determination of the ph of peri-implant human mouths involved in the production and Actinomyces naeslundii. Oral Microbiology and
crevicular fluid in successful and failing dental utilization of hydrogen peroxide. Archives of Immunology 11, 193–198.
implant sites: a pilot study. Archives of Oral Oral Biology 40, 753–763. Tan, K. H., Seers, C. A., Dashper, S. G., Mitch-
Biology 50, 1055–1059. Schincaglia, G. P., Hong, B. Y., Rosania, A., ell, H. L., Pyke, J. S., Meuric, V., Slakeski, N.,
Okuda, T., Kokubu, E., Kawana, T., Saito, A., Barasz, J., Thompson, A., Sobue, T., Pana- Cleal, S. M., Chambers, J. L., McConville, M.
Okuda, K. & Ishihara, K. (2012) Synergy in gakos, F., Burleson, J. A., Dongari-Bagtzoglou, J. & Reynolds, E. C. (2014) Porphyromonas
biofilm formation between Fusobacterium A. & Diaz, P. I. (2017) Clinical, immune, and gingivalis and Treponema denticola exhibit
nucleatum and Prevotella species. Anaerobe 18, microbiome traits of gingivitis and peri-implant metabolic symbioses. PLoS Pathogens 10,
110–116. mucositis. Journal of Dental Research 96, 47– e1003955.
Olczak, T., Simpson, W., Liu, X. & Genco, C. A. 55. Teughels, W., Durukan, A., Ozcelik, O., Pauwels,
(2005) Iron and heme utilization in Porphy- Scoffield, J. A. & Wu, H. (2015) Oral streptococci M., Quirynen, M. & Haytac, M. C. (2013)
romonas gingivalis. FEMS Microbiology and nitrite-mediated interference of Pseu- Clinical and microbiological effects of Lacto-
Reviews 29, 119–144. domonas aeruginosa. Infection and Immunity 83, bacillus reuteri probiotics in the treatment of
Olsen, I., Tribble, G. D., Fiehn, N.-E. & Wang, 101–107. chronic periodontitis: a randomized placebo-
B.-Y. (2013) Bacterial sex in dental plaque. Scoffield, J. A. & Wu, H. (2016) Nitrite reductase controlled study. Journal of Clinical Periodon-
Journal of Oral Microbiology 5, doi: 10.3402/ is critical for Pseudomonas aeruginosa survival tology 40, 1025–1035.
jom.v5i0.20736. during co-infection with the oral commensal Traudt, M. & Kleinberg, I. (1996) Stoichiometry
Park, O.-J., Yi, H., Jeon, J. H., Kang, S.-S., Koo, Streptococcus parasanguinis. Microbiology 162, of oxygen consumption and sugar, organic acid
K.-T., Kum, K.-Y., Chun, J., Yun, C.-H. & Han, 376–383. and amino acid utilization in salivary sediment
S. H. (2015) Pyrosequencing analysis of subgingi- Senadheera, D. & Cvitkovitch, D. G. (2008) Quo- and pure cultures of oral bacteria. Archives of
val microbiota in distinct periodontal conditions. rum sensing and biofilm formation by Strepto- Oral Biology 41, 965–978.
Journal of Dental Research 94, 921–927. coccus mutans. Advances in Experimental Vartoukian, S. R., Adamowska, A., Lawlor, M.,
Perez-Chaparro, P. J., Goncßalves, C., Figueiredo, Medicine and Biology 631, 178–188. Moazzez, R., Dewhirst, F. E. & Wade, W. G.
L. C., Faveri, M., Lob~ ao, E., Tamashiro, N., Sharma, A., Inagaki, S., Sigurdson, W. & Kura- (2016a) Cultivation of ‘unculturable’ oral bac-
Duarte, P. & Feres, M. (2014) Newly identified mitsu, H. K. (2005) Synergy between Tan- teria, facilitated by community culture and
pathogens associated with periodontitis: a sys- nerella forsythia and Fusobacterium nucleatum media supplementation with siderophores.
tematic review. Journal of Dental Research 93, in biofilm formation. Oral Microbiology and PLoS One 11, e0146926.
846–858. Immunology 20, 39–42. Vartoukian, S. R., Moazzez, R. V., Paster, B. J.,
Peterson, S. N., Snesrud, E., Liu, J., Ong, A. C., Shu, M., Browngardt, C. M., Chen, Y.-Y. M. & Dewhirst, F. E. & Wade, W. G. (2016b) First
Kilian, M., Schork, N. J. & Bretz, W. (2013) Burne, R. A. (2003) Role of urease enzymes in cultivation of health-associated Tannerella sp.
The dental plaque microbiome in health and stability of a 10-species oral biofilm consortium Hot-286 (BU063). Journal of Dental Research
disease. PLoS ONE 8, e58487. cultivated in a constant-depth film fermenter. 95, 1308–1313.
Petersen, F. C., Tao, L. & Scheie, A. A. (2005) Infection and Immunity 71, 7188–7192. Wade, W. G. (2013) The oral microbiome in
DNA binding-uptake system: a link between Sim on-Soro, A., Guillen-Navarro, M. & Mira, A. health and disease. Pharmacological Research
cell-to-cell communication and biofilm forma- (2014) Metatranscriptomics reveals overall 69, 137–143.
tion. Journal of Bacteriology 187, 4392–4400. active bacterial composition in caries lesions. Walker, C. B., Tyler, K. Z., Low, S. B. & King,
Poulsen, K., Reinholdt, J., Jespersgaard, C., Journal of Oral Microbiology 6, doi: 10.3402/ C. J. (1987) Penicillin-degrading enzymes in
Boye, K., Brown, T. A., Hauge, M. & Kilian, jom.v6.25443. sites associated with adult periodontitis. Oral
M. (1998) A comprehensive genetic study of Slaney, J. M. & Curtis, M. A. (2008) Mecha- Microbiology and Immunology 2, 129–131.
streptococcal immunoglobulin A1 proteases: nisms of evasion of complement by Porphy- Wang, B.-Y., Deutch, A., Hong, J. & Kuramitsu,
evidence for recombination within and between romonas gingivalis. Frontiers in Bioscience 13, H. K. (2011) Proteases of an early colonizer can
species. Infection and Immunity 66, 181–190. 188–196. hinder Streptococcus mutans colonization
Pride, D. T., Salzman, J., Haynes, M., Rohwer, F., Stacy, A., Everett, J., Jorth, P., Trivedi, U., Rum- in vitro. Journal of Dental Research 90, 501–505.
Davis-Long, C., White, R. A. III, Loomer, P., baugh, K. P. & Whiteley, M. (2014) Bacterial Wang, J., Gao, Y. & Zhao, F. (2016) Phage-bac-
Armitage, G. C. & Relman, D. A. (2012) Evi- fight-and-flight responses enhance virulence in teria interaction network in human oral micro-
dence of a robust resident bacteriophage popula- a polymicrobial infection. Proceedings of the biome. Environmental Microbiology 18, 2143–
tion revealed through analysis of the human National Academy of Sciences of the United 2158.
salivary virome. ISME Journal 6, 915–926. States of America 111, 7819–7824. Wei, G. X., van der Hoeven, J. S., Smalley, J. W.,
Rams, T. E., Degener, J. E. & van Winkelhoff, A. ter Steeg, P. F. & van der Hoeven, J. S. (1989) Mikx, F. H. & Fan, M. W. (1999) Proteolysis
J. (2013) Prevalence of b-lactamase-producing Development of periodontal microflora on and utilization of albumin by enrichment cul-
bacteria in human periodontitis. Journal of human serum. Microbial Ecology in Health and tures of subgingival microbiota. Oral Microbi-
Periodontal Research 48, 493–499. Disease 2, 1–10. ology and Immunology 14, 348–351.

© 2017 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd
S22 Marsh and Zaura

van Winkelhoff, A. J., Winkel, E. G., Barendregt, Zeng, L., Farivar, T. & Burne, R. A. (2016) Table S1. PubMed query search
D., Dellemijn-Kippuw, N., Stijne, A. & van der Amino sugars enhance the competitiveness of
terms and results.
Velden, U. (1997) Β-lactamase producing bac- beneficial commensals with Streptococcus
teria in adult periodontitis. Journal of Clinical mutans through multiple mechanisms. Applied
Periodontology 24, 538–543. and Environment Microbiology 82, 3671–3682.
Wright, C. J., Burns, L. H., Jack, A. A., Back, C. Zijnge, V., van Leeuwen, M. B. M., Degener, J.
R., Dutton, L. C., Nobbs, A. H., Lamont, R. E., Abbas, F., Thurnheer, T., Gm€ ur, R. &
J. & Jenkinson, H. F. (2013) Microbial interac- Harmsen, H. J. M. (2010) Oral biofilm
tions in building of communities. Molecular architecture on natural teeth. PLoS One 5,
Oral Microbiology 28, 83–101. e9321.
Xu, H., Jenkinson, H. F. & Dongari-Bagtzoglou, Address:
A. (2014) Innocent until proven guilty: mecha- E. Zaura
nisms and roles of Streptococcus-Candida inter- Department of Preventive Dentistry
actions in oral health and disease. Molecular Academic Centre for Dentistry Amsterdam
Oral Microbiology 29, 99–116. Supporting Information Gustav Mahlerlaan 3004
Yost, S., Duran-Pinedo, A. E., Teles, R., Krish-
nan, K. & Frias-Lopez, J. (2015) Functional Additional Supporting Information 1081 LA Amsterdam
signatures of oral dysbiosis during periodontitis may be found in the online version The Netherlands
progression revealed by microbial metatran- E-mail: e.zaura@acta.nl
scriptome analysis. Genome Medicine 7, 27.
of this article:

Clinical Relevance influence oral biofilm composition environmental perturbations, which


Scientific rationale for the study: and activity. contributes to oral health.
Microorganisms persist in the Principal findings: A literature review Practical implications: Treatment
mouth as multispecies biofilms confirmed that numerous synergistic strategies should also include
that deliver important benefits to and antagonistic interactions occur reducing environmental pressures
the host. Microbes will interact among the resident microbes, result- that drive dysbiosis so that a
because of their physical proxim- ing in tightly integrated communities favourable ecological balance is
ity, and the outcome will that are resilient against minor maintained.

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

You might also like