You are on page 1of 8

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/268883659

Solving the etiology of dental caries

Article  in  Trends in Microbiology · November 2014


DOI: 10.1016/j.tim.2014.10.010

CITATIONS READS

263 7,073

2 authors:

Aurea Simon-Soro Alex Mira


University of Pennsylvania FISABIO
36 PUBLICATIONS   1,749 CITATIONS    190 PUBLICATIONS   8,827 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

SIMMA test View project

The Oral Microbiome in Lung Transplant Recipients View project

All content following this page was uploaded by Alex Mira on 17 May 2018.

The user has requested enhancement of the downloaded file.


Opinion

Solving the etiology of dental caries


Aurea Simón-Soro and Alex Mira
FISABIO Foundation, Center for Advanced Research in Public Health, Avda Cataluña 21, 46020 Valencia, Spain

For decades, the sugar-fermenting, acidogenic species diseased subjects [11]. An important hurdle in determining
Streptococcus mutans has been considered the main the etiology of tooth decay is that many samples were not
causative agent of dental caries and most diagnostic taken from the disease site itself but from other, noninva-
and therapeutic strategies have been targeted toward sive samples such as saliva, which does not represent the
this microorganism. However, recent DNA- and RNA- cariogenic microbiota (Box 1). However, in a seminal work,
based studies from carious lesions have uncovered an Aas and collaborators obtained over 1200 clones of the 16S
extraordinarily diverse ecosystem where S. mutans rRNA gene from dental plaques and carious lesions at
accounts only a tiny fraction of the bacterial community. different stages of the disease [12]. This work showed that
This supports the concept that consortia formed by S. mutans could not be PCR amplified in a significant
multiple microorganisms act collectively, probably syn- proportion of samples and other bacteria such as Atopo-
ergistically, to initiate and expand the cavity. Thus, bium, Prevotella, and Propionibacterium appeared to be
antimicrobial therapies are not expected to be effective associated with the disease. Recent work added Scardovia
in the treatment of caries and other polymicrobial dis- wiggsiae as a new etiological agent of severe early child-
eases that do not follow classical Koch’s postulates. hood caries [13]. In recent years, the use of second-genera-
tion sequencing and metagenomic techniques has
Microbiology of dental caries uncovered an extraordinarily diverse ecosystem where
Classical Koch’s postulates contend that a specific micro- S. mutans accounts only for 0.1% of the bacterial commu-
organism can be found to be responsible for an infectious nity in dental plaque and 0.7–1.6% in carious lesions
disease when it invades a host, a principle that has been [14,15]. When the DNA of samples from dentin caries
assumed to be correct for most microbial infections. Al- was directly sequenced, obviating cloning or PCR techni-
though the identification of asymptomatic carriers readily ques, Veillonella appeared as the most common genus [16],
showed that the postulates have important limitations and underlining the varying nature of microbial composition in
the original formulation has been modified with the intro- cavities. However, these DNA-based studies may quantify
duction of genetic techniques [1], the principles proposed dead, transient, or inactive microorganisms that do not
by Koch have remained a cornerstone in microbiology. contribute to the disease, inflating estimates of diversity
Probably due to this, when the sugar-fermenting, acido- and introducing noise in the analysis [17]. Thus, the ap-
genic species S. mutans was isolated in the 1920s from plication of high-throughput sequencing to the RNA
carious lesions, it was considered to be the etiological agent extracted from oral samples finally provides an opportuni-
of dental caries [2]. Dental caries is considered the most ty to identify the metatranscriptome; that is, the active
prevalent human disease, affecting 80–90% of the world microbial composition and expressed genetic repertoire
population [3]. In children, it appears to have a fivefold underlying disease initiation and progression.
higher prevalence than asthma, which is the second most The first of these RNA-based studies on the surface of
prevalent disease. For decades, mutans streptococci have teeth [18] studied the active microbial communities in oral
been considered the main causative agent of the disease [4] biofilms before and after a meal, identifying the bacteria
and most diagnostic, preventive, and therapeutic strate- that increase their activity after food ingestion, with the
gies have been targeted toward this microorganism (see, premise that these organisms may be involved in sugar
for example, [5–7]). However, other microbial species were fermentation and acid formation. Metatranscriptomic data
also isolated from carious lesions and have been related indicate that the active microbiota is a subset of the total
to the process of tooth decay, including lactobacilli [8] microbial composition in oral biofilms [19] but is still
and bifidobacteria [9]. The introduction of molecular extraordinarily diverse. In addition, the RNA-based esti-
approaches to study the human microbiome revealed that mates of diversity indicate that different microbial consor-
the oral ecosystem is inhabited by hundreds of bacterial tia are formed in the dental plaque of different individuals.
species [10], most of which are considered commensals, and Thus, determining the active microbiota in carious lesions
that species regarded as pathogens are frequently found may finally unravel the elusive etiology of the disease,
in healthy individuals, although at lower levels than in paving the way for diagnostic and preventive tools.

Corresponding author: Mira, A. (mira_ale@gva.es).


The active microbiota of cavities
Keywords: metatranscriptomics; tooth decay; dysbiosis; Koch’s postulates; patho-
biont. The first RNA-based estimates of bacterial diversity in
0966-842X/
cavities are shown in Figure 1, putatively representing
ß 2014 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tim.2014.10.010 the microbial consortia that are actively contributing to the
disease. This approach shows an average of eight active
76 Trends in Microbiology, February 2015, Vol. 23, No. 2
Opinion Trends in Microbiology February 2015, Vol. 23, No. 2

genera at a presence of over 1% in both enamel and dentin [25]. For instance, S. gordonii modulates biofilm formation
lesions. However, each lesion appears to harbor a different in Candida albicans and Candida load influenced oral
combination of bacteria. There was only one case in which bacterial diversity and antibiotic resistance (reviewed in
the lesion was dominated by a single bacterial genus [26]). Also, viruses may play a vital role in shaping microbial
(marked as sample CA085, tooth 47 in the figure). This populations [27], but this has been poorly studied in oral
exceptional instance involved Lactobacillus, which was environments [27].
found to represent 99% of the RNA-based population in In the putative list of caries-associated bacteria
a hidden dentin cavity (55% of the reads had maximal revealed by this metatranscriptomic approach, S. mutans
similarity to Lactobacillus gasseri, but three other species accounts for 0.02% of the active microorganisms in hidden
of lactobacilli were present). However, all of the other dentin cavities, 0.48% in open dentin cavities, and 0.73% in
samples showed diverse bacterial compositions that varied enamel carious lesions [20]. Thus, although there is sub-
dramatically between individuals, even within the same stantial evidence that S. mutans is associated with caries
carious lesion type (Figure 1). The estimated bacterial risk, other species clearly arise as main players in the
diversity was lowest for enamel carious lesions, with an microbial community, including Veillonella, Rothia, and
average of 177 species-level phylotypes per sample, and Leptotrichia in enamel caries and Streptococcus sanguinis,
highest for ‘open’ dentin cavities that were exposed to the Atopobium, Schlegelella, Pseudoramibacter, and Lactoba-
oral cavity, which had an average of 251 species per cillus in dentin caries. Some of these bacteria are poorly
sample. This suggests that open dentin cavities have an characterized, as exemplified by the genus Schlegelella, in
input of microorganisms from saliva, even if the biofilm which the 16S rRNA sequences identified in this and other
formed on top of the lesion is eliminated from the sample, studies [28] indicate that this caries-associated oral mi-
as was the case here. ‘Hidden’ dentin cavities, which have crobe is a different species from the only two isolated
almost no contact with the oral cavity except for a minimal organisms in this genus that are currently described, both
lesion through the enamel, had an estimated number of in nonhuman niches. The polymicrobial nature of carious
species-level phylotypes of 201 [20]. The observation that lesions implies that animal models are probably not rep-
multiple species are detected by their RNA in carious resentative of human oral disease, especially in cases
lesions unequivocally demonstrates that they are alive where single bacterial species are inoculated in the animal
and supports the concept that consortia formed by multiple [29,30].
microorganisms act collectively to initiate and expand the A revealing aspect of RNA-based studies is that the
cavity. It has been shown that several oral species can act composition of active bacteria in initial, enamel lesions
synergistically to increase their pathogenic effect [21]. In a appears to be different from that found in more advanced
fascinating example of metabolite cross-feeding, Strepto- dentin cavities. This observation holds even in cases where
coccus gordonii was shown to increase the virulence of enamel and dentin cavities from the same tooth were
Aggregatibacter actinomycetemcomitans in an animal ab- sampled and analyzed. An example is shown in
scess model [22]. Mixed cultures of S. mutans and Veillo- Figure 2, where the hidden dentin cavity of a molar tooth
nella alcalescens were also found to produce higher acid had high frequencies of Neisseria, Lactobacillus, Mega-
levels than biofilms containing only one of these species sphaera, and Rothia, whereas a non-cavitated enamel
[23]. In addition, Veillonella may favor acid-producing bac- caries lesion had high frequencies of Haemophilus and
teria in caries through nitrate reduction [24], given that low Gemella. In addition, the streptococci were dominated by
concentrations of nitrite killed several cariogenic organ- S. sanguinis in the dentin cavity, while Streptococcus mitis
isms. Finally, it must be remembered that not only bacterial was at significantly higher levels in the enamel caries
interactions but also bacterial–fungal associations can be lesion, which also showed a higher streptococcal diversity
vital for promoting virulence in disease-associated consortia [20]. A previous metagenomic study demonstrated that

Box 1. Targeting the causative agents of dental caries


DNA-based studies of microbial diversity in the oral cavity have instance, [54,55]). However, when saliva, dental plaque, and carious
estimated that the human supragingival dental plaque (the biofilm lesions from the same individuals are analyzed, it is readily observed
formed on tooth surfaces) contains between 500 and 700 bacterial that saliva is not representative of the bacterial diversity located at the
species [10,12,16]. These estimates are even higher in saliva, probably disease site (Figure I). The microbial composition of the enamel lesion
because this oral fluid is in contact with all niches in the mouth, under study (orange circle) appears to be dominated by Veillonella,
reaching values between 1000 and 2000 species in stimulated saliva Fusobacterium, and Porphyromonas, whereas in the saliva sample
[52]. In carious lesions, however, the number decreases dramatically (blue, outer circle) from the same individual Streptococcus, Neisseria,
to 100–200 species-level phylotypes, both in initial, enamel caries and Prevotella are the genera found at the highest proportions. This is
lesions and in dentin or deep-dentin cavities [14,15,35,53], but in agreement with other studies that strongly suggest that saliva
because these studies are based on PCR amplification of DNA many samples are not appropriate for studying the microbiology of oral
of the organisms detected may be inactive and not contributing to diseases [52,56]. Although the dental plaque samples (green circle)
lesion progression. The recent RNA-based data (see Figure 1 in main are more similar in bacterial composition to that found in their
text) identify bacteria that are actively involved in translation respective carious lesions, several genera decrease and other
processes, narrowing the list of caries-related organisms to 40– increase in proportion in the cavity, as a consequence of the more
160 per sample, which are presumably those active in individual specialized niche. Thus, even dental plaque will not accurately show
cavities. It has been assumed for years that the bacteria involved in the bacterial communities responsible for dental caries and the data
the disease should also be present in saliva, which has been the strongly recommend the use of carious lesion samples with RNA-
preferred oral sample collected in etiological and epidemiological based approaches (pink, inner circle) to determine the active
studies of dental caries due to its noninvasive nature (see, for etiological agents of the disease.

77
Opinion Trends in Microbiology February 2015, Vol. 23, No. 2

TRENDS in Microbiology

Figure I. Bacterial composition of different oral samples from the same individual. Each ring corresponds to DNA extracted from drooling saliva, dental plaque from
sound surfaces, and an enamel carious lesion, as well as from RNA from the same lesion (inner circle). Font size is related to the proportion of each taxonomic group in
the sample.

enamel caries bacteria have an over-representation of and osmotic stress genes in dentin cavities [14]. Thus, the
dietary sugar-fermenting genes, whereas dentin caries data clearly demonstrate that caries microbiology is tissue
organisms are enriched in genes involved in the metabo- dependent and does not a have a unique etiology, which
lism of human-associated glycans [14]. In addition, enamel has important consequences for disease prevention.
caries microorganisms are extremely rich in adhesion
molecules whereas the microbial community in dentin Preventing tooth decay
caries contains a remarkable arsenal of proteases to de- The fact that caries-associated bacterial consortia vary at
grade dentinary tissue, including collagenases, dipeptidyl different stages of disease progression and are dissimilar
peptidases, serine proteases, glycoproteases, matrix metal- in different individuals suggests that the same functions
lopeptidases, and aminopeptidases. The environments may be performed by different sets of microorganisms. This
that the two cariogenic microbial consortia inhabit are also appears to be a general trend in metagenomic studies of the
reflected in the most common stress genes: oxidative stress human microbiome, where radically different taxonomic
genes in both tissues, acidic stress genes in enamel lesions, compositions give rise to surprisingly similar functional
78
Opinion Trends in Microbiology February 2015, Vol. 23, No. 2

100%
Key: Other
Veillonella
90% Streptococcus
Selenomonas
80% Schlegelella
Rothia
Pseudoramibacter
70% Prevotella
Porphyromonas
60% Neisseria
Leptotrichia
Lactobacillus
50% Kingella
Granulicatella
40% Gemella
Fusobacterium

30% Eubacterium
Corynebacterium

20% Capnocytophaga
Atopobium
Acnomyces
10% Abiotrophia

0%
CA 1.3 S
CA 21.4 WS
CA 22.1 WS
CA 22.4 WS
CA 2.4 S
CA 78.3 WS
CA 78.3 WS
CA 78.4 WS
CA 8.4 S
CA 8.4 WS
CA 8.4 S
CA 89.4 WS
CA 01.1 WS
1. S
S

CA 77.4 DD
CA 79.1 DD
CA 79.2 DD
CA 82.3 DD
8. D
DD

CA 78.4 H
08 7H

CA 83.2 H
CA 85.4 H
08 7H
H
02 4W

02 6W

07 5W

08 7W

10 2W
W

08 4D

0 7

0 8
0 7

28
CA 8.3

CA 1.4
0 8
0 8
0 5
0 1

46
0 6
0 6
0 3

0 7
0 6
0 7

07 6

0 7
1 5

23

6.
CA 7.3
CA 21.3

07
07

CA
0

CA
CA

Samples
TRENDS in Microbiology

Figure 1. Metatranscriptomic profile of caries lesions. RNA was extracted from initial, white-spot enamel caries (WS), the deep layer from open dentin cavities (DD), and
hidden dentin cavities (H). After cDNA construction, bacterial taxonomy was obtained by Titanium-plus pyrosequencing of 800 bp PCR products encompassing
hypervariable regions V1 through V4 and using the Ribosomal Database Project (RDP) classifier. Over 4000 reads per sample were obtained, giving information about the
active microbial community. The two numbers to the right of the patient code indicate the tooth from where the sample was obtained. Drawn from data in [20].

profiles [31]. Thus, even if determining the etiology of a in predicting risk of oral diseases and could have poten-
microbe-mediated disease could be considered the first tial diagnostic value.
step in designing preventive strategies, knowing the Thus, the change in paradigm in the etiology of tooth
specific microbial composition will not suffice to under- decay must be translated to appropriate therapies
stand and combat the disease. Initial hypotheses about [35]. Given the polymicrobial nature of dental caries, we
the underlying microbiology of oral diseases, which fo- predict that diagnostic and preventive strategies directed
cused on determining the specific causative agents, gave toward specific bacterial species will not be universally
way to ecology-based propositions where the disease is effective. These include diagnostic kits of caries risk as-
seen as the output of a skewed microbial community due sessment, which are traditionally focused on culture
to environmental changes [32]. If the microbial composi- counts of mutans streptococci and lactobacilli [4] whereas
tion of carious lesions is so variable and the combinations the data show that the former are found at extremely low
of possible consortia so numerous (Figure 1), is it relevant proportions even within cavities and the latter are found
to know a list of cariogenic players? The ecological hy- only in several dentin cavities and not in initial, enamel
pothesis of dental caries considers that focusing on the caries lesions (Figure 1), confirming that this species is not
metabolic output of microbial communities can be the involved in the initiation of caries [36]. Regarding preven-
clue to understanding and controlling the disease, re- tive or therapeutic approaches, passive immunization
gardless of the microorganisms involved in the process strategies such as the use of synthetic antibodies directed
[33], and different omic approaches have been initiated to against S. mutans-specific antigens [37] are predicted to be
understand the holistic functional output of dental pla- inefficient given the multispecies nature of caries initia-
que [17,34]. From an applied viewpoint, we believe that tion. Active immunization strategies (i.e., a caries vaccine)
the detection of disease-associated metabolic profiles are being explored mainly against mutans streptococci
would be more informative than microbial composition antigens [6,38] and even the multispecies approach that
79
Opinion Trends in Microbiology February 2015, Vol. 23, No. 2

Atopobium
erium
Fusobact

us
a
ll
phil

ge
Kin
mo

s
ilu
ra

ac
Hae
ae
sph

ob
ega

ct
M
Veill

La
onel
la
Neis
seria
Stre
pto
coc
CA078.47H cus

Por
Sa

Rot
phi
gu

ro
n

hia
mo
inis

nas
Mis
Gemella

us
hil
op
em
Ha

Veill
onel
la
us
o cc
t oc
Mis

ep
Str

San
g ui n is
CA078.47WS

TRENDS in Microbiology

Figure 2. The bacterial composition of caries is tissue dependent. A sample from an initial, white-spot enamel caries (WS) and a hidden dentin lesion were taken from the
same tooth (the second molar from the fourth quadrant). The lack of contact between the two lesions was assessed radiographically. The active, RNA-based bacterial
taxonomy was obtained by pyrosequencing of long PCR products encompassing hypervariable regions V1 through V4. Drawn from data in [20].

was later proposed [39] may not suffice if a key, universal


bacterial repertoire is not present in caries development.
Thus, future research should focus on intercepting func- Box 2. Outstanding questions
tions related to disease initiation and progression regard-
 Is the functional output of a microbial community more important
less of which is the bacterial player behind the role (Box 2). than its taxonomic composition in terms of understanding a
Tentative examples of functions to be inhibited in future polymicrobial disease?
caries preventive strategies might include quorum sensing  Are immunization and antimicrobial strategies effective in pre-
and other intercellular communication signals, carbohy- venting oral diseases?
 Are we treating polymicrobial oral disorders as infectious single-
drate-fermenting pathways, intermicrobial and microbial– species diseases?
human adhesion compounds, bacterial–immune system  What non-antimicrobial therapeutic strategies are feasible to
crosstalk molecules, and pH-regulating particles. Disrup- prevent polymicrobial diseases such as dental caries?
tion of adhesion molecules may be a fruitful strategy to  Given that the human microbiome contains not only bacteria but
also fungal and viral organisms, how common are polymicrobial
either prevent or modulate microbial attachment to the
(as opposed to polybacterial) diseases?
tooth pellicle or between key players in biofilm formation
80
Opinion Trends in Microbiology February 2015, Vol. 23, No. 2

Table 1. Key features of single-species and polymicrobial oral diseases


Etiology Type of disease Present in health Virulent behavior Immune response Treatment
Single species Infectious No (invasive) Pathogenic Active Antimicrobial
Polymicrobial Dysbiosis Yes (commensal) Pathobiont Basal Restore balance

[40]. This is based on the premise that interference in the polymicrobial diseases is emerging outside the oral cavity,
development of the oral biofilm caused by impairing the ranging from abscesses to bacterial vaginosis, diarrhea,
tooth attachment of early colonizers or their ability to rhinosinusitis, and chronic infection in the lung of patients
recruit other key players during biofilm formation would with cystic fibrosis [49]. The common detection of patho-
affect the entire process and avoid the presence of cario- gens present at low levels in health in most polymicrobial
genic or periodontal pathogens. For instance, Fusobacter- oral diseases strongly suggests that they cannot be consid-
ium nucleatum is a promising candidate given its ered to have an infectious nature and that the microbial
capability to coaggregate with both early and late coloni- causative agents are better described as pathobionts
zers of the oral biofilm [41]. Controlled (e.g., at subinhibi- [50,51]. These organisms are therefore resident bacteria
tory levels), limited, or specific use of antibiotic therapy has with the potential to cause disease, and under balanced
also been proposed [42]. conditions the immune system does not have an active
Another alternative to modulate plaque development response against them (Table 1). From an applied view-
without making use of an antimicrobial strategy is the point, we believe that antimicrobial treatments will be
use of probiotics. The introduction of strains specific to unfruitful in healing or preventing oral polymicrobial dis-
the biofilm could theoretically modify key microbial inter- eases and strategies directed toward modulating microbial
actions or buffer cariogenic effects, favoring a healthy, bal- interactions and/or their functional output should be fur-
anced ecosystem. However, current clinical trials with ther developed.
probiotic treatment have not been particularly effective in
reducing caries rates [43]. A reason for this can be related to References
the use of single species in probiotic approaches. Not only 1 Fredericks, D.N. and Relman, D.A. (1996) Sequence-based
identification of microbial pathogens: a reconsideration of Koch’s
pathogens act in groups: balance-promoting commensals
postulates. Clin. Microbiol. Rev. 9, 18–33
probably need multispecies clusters to their facilitate set- 2 Loesche, W.J. (1986) Role of Streptococcus mutans in human dental
tling on the biofilm and to provide beneficial effects. Perhaps decay. Microbiol. Rev. 50, 353–380
for this reason, in the human gut, fecal transplantations 3 Petersen, P.E. (2004) Challenges to improvement of oral health in the
have been shown to be effective in treating Clostridium 21st century – the approach of the WHO Global Oral Health
Programme. Int. Dent. J. 54, 329–343
difficile infections, colitis, and irritable bowel syndrome,
4 Loesche, W.J. et al. (1975) Association of Streptococcus mutants with
suggesting that an already-formed community settles more human dental decay. Infect. Immun. 11, 1252–1260
effectively in the recipient niche and has a higher probability 5 Plonka, K.A. et al. (2012) Mutans streptococci and lactobacilli
of conferring a positive effect [44]. However, the practical colonization in predentate children from the neonatal period to
application of oral transplants would have important draw- seven months of age. Caries Res. 46, 213–220
6 Kt, S. et al. (2013) Dental caries vaccine – a possible option? J. Clin.
backs in daily use, ranging from lack of reproducibility to Diagn. Res. 7, 1250–1253
social rejection of patients or lack of control in the dose and 7 Islam, B. et al. (2007) Dental caries: from infection to prevention. Med.
composition of the administered material. A potential solu- Sci. Monit. 13, RA196–RA203
tion could be the identification of microbial species that may 8 Badet, C. and Thebaud, N.B. (2008) Ecology of lactobacilli in the oral
form stable, cooperative consortia in healthy individuals cavity: a review of literature. Open Microbiol. J. 2, 38–48
9 Mantzourani, M. et al. (2009) Association between Bifidobacteriaceae
and that could be used to design probiotic complexes with and the clinical severity of root caries lesions. Oral Microbiol. Immunol.
preventive purposes. In addition to these controlled oral 24, 32–37
transplants, prebiotics – substances that promote the 10 Bik, E.M. et al. (2010) Bacterial diversity in the oral cavity of ten
growth of specific microbial consortia – are a promising healthy individuals. ISME J. 4, 962–974
11 Human Microbiome Project Consortium (2012) Structure, function and
option but have unfortunately been only modestly explored
diversity of the healthy human microbiome. Nature 486, 207–214
[45]. Future metatranscriptomic, proteomic, or metabolomic 12 Aas, J.A. et al. (2008) Bacteria of dental caries in primary and
approaches [46] may provide clues to identify key molecules permanent teeth in children and young adults. J. Clin. Microbiol.
that could interfere with dental caries initiation and pro- 46, 1407–1417
gression [17] and could circumvent the complex and elusive 13 Tanner, A.C. et al. (2011) Cultivable anaerobic microbiota of severe
early childhood caries. J. Clin. Microbiol. 49, 1464–1474
polymicrobial nature of this and other oral diseases.
14 Simón-Soro, A. et al. (2013) A tissue-dependent hypothesis of dental
caries. Caries Res. 47, 591–600
Polymicrobial versus infectious disease 15 Gross, E.L. et al. (2012) Beyond Streptococcus mutans: dental caries
Apart from dental caries, other oral diseases have also onset linked to multiple species by 16S rRNA community analysis.
been clearly identified as polymicrobial, including gingivi- PLoS ONE 7, e47722
16 Belda-Ferre, P. et al. (2012) The oral metagenome in health and
tis, halitosis, and root canal infections [47]. A well-studied disease. ISME J. 6, 46–56
case is periodontitis, where even the initial three-species 17 Nyvad, B. et al. (2013) Dental caries in a molecular microbiological
cluster proposed to underlie disease development (the so- perspective. Caries Res. 47, 89–102
called ‘red complex’ of periodontal disease) has proved to be 18 Benı́tez-Páez, A. et al. (2014) Microbiota diversity and gene expression
insufficient to explain its etiology [48]. An expanding list of dynamics in the human oral biofilm. BMC Genomics 15, 311

81
Opinion Trends in Microbiology February 2015, Vol. 23, No. 2

19 Duran-Pinedo, A.E. et al. (2014) Community-wide transcriptome of the 38 Zhang, S. (2014) Dental caries and vaccination strategy against the
oral microbiome in subjects with and without periodontitis. ISME J. 8, major cariogenic pathogen, Streptococcus mutans. Curr. Pharm.
1659–1672 Biotechnol. 14, 960–966
20 Simón-Soro, A. et al. (2014) Metatranscriptomics reveals active 39 Mira, A. (2007) Horizontal gene transfer in oral bacteria. In Oral
bacterial composition in caries lesions. J. Oral Microbiol. 6, 25443 Molecular Microbiology (Rogers, A.H., ed.), pp. 65–86, Horizon
21 Murray, J.L. et al. (2014) Mechanisms of synergy in polymicrobial Scientific Press
infections. J. Microbiol. 52, 188–199 40 Wright, C.J. et al. (2014) Disruption of heterotypic community
22 Ramsey, M.M. et al. (2011) Metabolite cross-feeding enhances development by Porphyromonas gingivalis with small molecule
virulence in a model polymicrobial infection. PLoS Pathog. 7, e1002012 inhibitors. Mol. Oral Microbiol. 29, 185–193
23 Noorda, W.D. et al. (1998) Monobacterial and mixed bacterial plaques 41 Kolenbrander, P.E. et al. (2010) Oral multispecies biofilm development
of Streptococcus mutans and Veillonella alcalescens in an artificial and the key role of cell–cell distance. Nat. Rev. Microbiol. 8, 471–480
mouth: development, metabolism, and effect on human dental 42 Rogers, G.B. et al. (2010) Revealing the dynamics of polymicrobial
enamel. Caries Res. 22, 342–347 infections: implications for antibiotic therapy. Trends Microbiol. 18,
24 Doel, J.J. et al. (2005) Evaluation of bacterial nitrate reduction in the 357–364
human oral cavity. Eur. J. Oral Sci. 113, 14–19 43 Cagetti, M.G. et al. (2013) The use of probiotic strains in caries
25 Harriott, M.M. and Noverr, M.C. (2011) Importance of Candida– prevention: a systematic review. Nutrients 5, 2530–2550
bacterial polymicrobial biofilms in disease. Trends Microbiol. 19, 44 Austin, M. et al. (2014) Fecal microbiota transplantation in the
557–563 treatment of Clostridium difficile infections. Am. J. Med. 127, 479–483
26 Krom, B.P. et al. (2014) Candida and other fungal species: forgotten 45 Devine, D.A. and Marsh, P.D. (2009) Prospects for the development of
players of healthy oral microbiota. J. Dent. Res. 93, 445–451 probiotics and prebiotics for oral applications. J. Oral Microbiol. 1, 1
27 Pride, D.T. et al. (2012) Evidence of a robust resident bacteriophage 46 Hart, T.C. et al. (2011) Identification of microbial and proteomic
population revealed through analysis of the human salivary virome. biomarkers in early childhood caries. Int. J. Dent. 2011, 196721
ISME J. 6, 915–926 47 Rôças, I.N. and Siqueira, J.F., Jr (2012) Characterization of microbiota
28 Lim, S.M. et al. (2011) Microbial profile of asymptomatic and of root canal-treated teeth with posttreatment disease. J. Clin.
symptomatic teeth with primary endodontic infections by Microbiol. 50, 1721–1724
pyrosequencing. J. Korean Acad. Conserv. Dent. 36, 498–505 48 Hajishengallis, G. and Lamont, R.J. (2012) Beyond the red complex
29 Xu, J.S. et al. (2014) Effect of emodin on the cariogenic properties of and into more complexity: the polymicrobial synergy and dysbiosis
Streptococcus mutans and the development of caries in rats. Exp. Ther. (PSD) model of periodontal disease etiology. Mol. Oral Microbiol. 27,
Med. 8, 1308–1312 409–419
30 Pekkala, E. et al. (2002) The effect of a high-sucrose diet on dentin 49 Peters, B.M. et al. (2012) Polymicrobial interactions: impact on
formation and dental caries in hyperinsulinemic rats. J. Dent. Res. 81, pathogenesis and human disease. Clin. Microbiol. Rev. 25, 193–213
536–540 50 Chow, J. and Mazmanian, S.K. (2010) A pathobiont of the microbiota
31 Vaishampayan, P.A. et al. (2010) Comparative metagenomics and balances host colonization and intestinal inflammation. Cell Host
population dynamics of the gut microbiota in mother and infant. Microbe 7, 265–276
Genome Biol. Evol. 2, 53–66 51 Ayres, J.S. et al. (2012) Lethal inflammasome activation by a
32 Marsh, P.D. (2003) Are dental diseases examples of ecological multidrug-resistant pathobiont upon antibiotic disruption of the
catastrophes? Microbiology 149, 279–294 microbiota. Nat. Med. 18, 799–806
33 Takahashi, N. and Nyvad, B. (2011) The role of bacteria in the caries 52 Simón-Soro, A. et al. (2013) Microbial geography of the oral cavity. J.
process: ecological perspectives. J. Dent. Res. 90, 294–303 Dent. Res. 92, 616–621
34 Takahashi, N. et al. (2010) Metabolomics of supragingival plaque and 53 Kianoush, N. et al. (2014) Bacterial profile of dentine caries and the
oral bacteria. J. Dent. Res. 89, 1383–1388 impact of pH on bacterial population diversity. PLoS ONE 9, e92940
35 Fejerskov, O. (2004) Changing paradigms in concepts on dental caries: 54 Nasidze, I. et al. (2009) Global diversity in the human salivary
consequences for oral health care. Caries Res. 38, 182–191 microbiome. Genome Res. 19, 636–643
36 Jiang, W. et al. (2014) Pyrosequencing analysis of oral microbiota 55 Rudney, J.D. et al. (2009) Potential biomarkers of human salivary
shifting in various caries states in childhood. Microb. Ecol. 67, 962–969 function: a modified proteomic approach. Arch. Oral Biol. 54, 91–100
37 Ma, J.K. (1998) Characterization of a recombinant plant monoclonal 56 Ling, Z. et al. (2010) Analysis of oral microbiota in children with dental
secretory antibody and preventive immunotherapy in human. Nat. caries by PCR-DGGE and barcoded pyrosequencing. Microb. Ecol. 60,
Med. 4, 601–606 677–690

82
View publication stats

You might also like