Periodontology 2000 - 2020 - Duran Pinedo - Metatranscriptomic Analyses of The Oral Microbiome

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

DOI: 10.1111/prd.

12350

REVIEW ARTICLE

Metatranscriptomic analyses of the oral microbiome

Ana E. Duran-Pinedo
Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA

Correspondence
Ana E. Duran-Pinedo, Department of Oral Biology, College of Dentistry, University of Florida, 1395 Center Dr., P.O. Box 100424, Gainesville, FL 32610, USA.
Email: aduran-pinedo@dental.ufl.edu

Funding information
National Institute of Dental and Craniofacial Research; National Institutes of Health, Grant/Award Number: 2R01DE021553

1 | I NTRO D U C TI O N a particular genus are detected. Thus, it is clear that we cannot rely
only on oral human studies because we need to compare the fre-
It is well known that the human body harbors trillions of microbes, quency and association of bacteria in oral sites with those in extra-
including bacteria, fungi, and viruses. A recent revision of the num- oral sites to establish a specific oral-related species.9,13-18 As a result
ber of bacteria residing in the human body puts the total number of the pioneering work of scientists such as F. Dewhirst, B. Paster, S.
of bacteria in a 70 kg "reference man" as 3.8 × 1013.1 The Human Socransky, and A. Hafajjee, among others, we are fortunate to have
Microbiome Project, an initiative of the National Institutes of Health solid knowledge on the specific composition of the indigenous flora
Road map for Biomedical Research (http://nihro​admap.nih.gov), in- in the oral cavity.
creased our understanding of microbial diversity in humans and its The golden era of the oral microbiome started 4 decades ago, in
role in human health and disease, as well as the dynamic interactions 1968, with the discovery of a link between bacteria as a causative
of microbes with host cells. Joshua Lederberg, in 2001, coined the agent of periodontal disease and caries. Worldwide, periodontitis
term microbiome "to signify the ecological community of commen- and caries are still the leading causes of tooth decay in adults, and
sal, symbiotic, and pathogenic microorganisms that share our body those with such diseases experience a marked decrease their quality
space and have been all but ignored as determinants of health and of life. Regarding periodontal diseases, the most recent data, from
2
disease." The term microbiome describes the whole community as a the National Health and Nutrition Examination Survey 2009-2014,19
"superorganism" in the context of their habitats. indicates that the prevalence of chronic periodontitis among adult
The “oral microbiome” comprises the bacteria, fungi,3 viruses,4-6 Americans is 42%, representing $14B expenditure on treatment in
archaea,7 and protozoa8 that inhabit the human oral cavity. This re- the US, and that periodontitis affects 743 million people worldwide.
view will focus on the bacterial part of the oral microbiome, and to Moreover, an increasing number of studies suggest that periodontal
simplify matters for the reader, we will subsequently refer to the diseases can influence the risk for certain systemic conditions, such
bacterial part as the oral microbiome. The oral microbiome is com- as cardiovascular diseases, diabetes, and respiratory diseases, and
posed of more than 700 species of bacteria9 with no significant dif- can affect pregnancy outcome. In a systematic analysis, Kassebaum
ference based on gender, race, age, or geographic location of host. et al reported a prevalence of untreated caries in permanent teeth of
However, the oral microbiome is distinct from that of the gut, colon, 34.1%, affecting 2.5 billion people worldwide. 20
10
or skin. The communities of the oral microbiome can be classified Current concepts of periodontal disease and caries etiology
as indigenous or exogenous (which is also known as the transient highlight the importance of oral biofilms as multispecies communi-
microbiome). The indigenous communities contain the oral bacte- ties. The focus has shifted from the role of individual species within
rial species found in virtually all human adults. On the other hand, the biofilm to a more holistic view of how biofilms, as a whole, cause
the transient exogenous communities include bacteria that are and maintain chronic infections. The metabolism of a bacterium is
present in the oral cavity, but whose natural niche is other than the modulated by its environment and is modified by the bacteria to
11,12
mouth. Differentiation between transient species and endoge- which it is in close proximity. The functions of distinct species within
nous species should be based on the frequency with which clones of the oral microbiota are intimately intertwined with the rest of the

© 2020 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

|
28     
wileyonlinelibrary.com/journal/prd Periodontology 2000. 2021;85:28–45.
DURAN-PINEDO |
      29

microbial community, highlighting the relevance of examining the mediators.39 The variety of ecological niches provided by the soft
expression profile of specific species of the oral microbiota in the and hard tissues within the mouth also determine the diversity and
context of the other members of the biofilm in vivo, as well as the abundance of the oral microbiome.40 The tooth surface close to the
expression pattern of the whole community. soft gingival tissue furnishes 2 major ecological niches: the suprag-
However, we should not dismiss the importance of studying indi- ingival area and the subgingival area. The supragingival area is ex-
vidual species. Knowledge of the function of bacterial species within posed to saliva, liquid, and solid food. The subgingival area is bathed
a community provides more insight into pathogenesis than the sim- by the serum transudate (known as gingival crevicular fluid), a fluid
ple enumeration of that community's gene content. rich in nutrients and proteins involved in the immune response.40
Nowadays, as a result of the development of next-generation se- The main difference between these 2 niches is the lower redox po-
quencing techniques for DNA and RNA sequencing and other omics tential, and the lower level of oxygen, in the subgingival area.41 Even
approaches, we can address those questions and obtain in situ in- though the environmental conditions are different, it is not easy to
formation on the shift in microbial community structure and met- make a statistical distinction between supragingival and subgingival
abolic activities that occur in the transition from health to disease. samples because they harbor almost the same group of microbes but
Among the powerful omics approaches is metatranscriptomics. The at different proportions.13,42-46
metatranscriptome refers to the expression of genes in the whole Moreover, the intermixing within the oral cavity is influenced by
microbial community. 21 Metatranscriptomics is more direct than saliva that coats all the oral surfaces. Bacteria transported by sa-
phylogenetic analysis. In contrast to 16S ribosomal RNA profiling, liva could fit into cracks or crevices, building biofilms in areas far
metatranscriptomic analysis provides information about the species from their site of origin. Besides, during sampling procedures (using
that are metabolically active in a community during health or disease a swab, buccal brush, paper point, or scaler), microbes can become
states. mixed in the biomass being sampled.47
Metatranscriptomic approaches have been successfully ap- As a result of limitations in the ability to discriminate species
22,23
plied to study diverse environmental microbial communities, using 16S ribosomal RNA analysis, the vast majority of studies have
and the use of this approach to study human-associated microbi- identified oral bacteria only to the level of genus.36 Nonetheless,
24-34
omes, such as periodontal diseases and caries, is increasing. given the significant variations in the structure and composition of
Metatranscriptomic analysis can answer crucial questions, such genomes belonging to the same species, in the future, we should aim
as: “What is the gene-expression profile of specific bacteria and to characterize the oral microbiome not even to species level but to
the whole community?”; “What are the functions of the expressed strain level, which will be accomplished with improvements both in
genes?”; “Which bacterial groups are expressing genes associated sequencing capabilities as well as in bioinformatics analysis.
with the types of activity observed?”; and “Are the genes expressed Supragingival and subgingival plaque harbor 14 common gen-
related to known virulence factors?” era: Streptococcus, Corynebacterium, Capnocytophaga, Haemophilus,
Moreover, metatranscriptomic gives us good insight into puta- Aggregatibacter, Fusobacterium, Prevotella, Leptotrichia, Veillonella,
tive factors assumed to drive the community to a disease state. That Neisseria, Rothia, Actinomyces, Lautropia, and Porphyromonas.
knowledge could also be applied to the development of new ther- Our current knowledge of the diversity of the oral microbiome
apies to maintain a healthy oral microbiome. Metatranscriptomic has been beautifully complemented with the direct observation of
analysis can also be applied to large sets of samples in longitudinal their structures, which was made possible by pioneer scientists in
studies and thus will provide a wealth of knowledge on populations. this field (J. M. Welch, A. Valm, and G. Borisy). They combined light
Moreover, the usefulness of metatranscriptomic approaches is not and electron microscopy to visualize the intact dental biofilm and
limited to the study of bacterial gene-expression profiles; it could unveiled the structure and organization of the oral community, with
also be applied to the host, to determine the pathways of bacte- no random distributions of polymicrobial biofilms.48-51
ria-host communication. Welch et al elegantly revealed the in vivo architecture of sam-
ples of supragingival dental plaque from healthy volunteers. By com-
bined use of fluorescence microscopy, antibodies, or oligonucleotide
2 |  CO M P OS ITI O N O F TH E O R A L probes52-55 and the multiplex fluorescence in-situ hybridization
M I C RO B I O M E technique, they visualized the 3-dimensional structure of undis-
turbed regions of supragingival biofilms.56 Large circular "hedgehog”
The healthy oral human microbiome is predominantly composed of and "corncob" structures were repeatedly observed. Bacteria of the
members of the phyla Actinobacteria, Proteobacteria, Firmicutes, genus Fusobacterium were at the core of the hedgehog arrangement
Bacteroidetes, and Fusobacter and Spirochaeteae are present at and were surrounded by bacteria from the genera Streptococcus,
lower number.13,35-38 The composition of the oral community is Porphyromonas, Leptotrichia, Capnocytophaga, and Actinomyces, and
shaped by physical and chemical interactions, environmental pres- from the families Pasturellaceae and Neisseriaceae. The corncob
sures (such as oxygen tension, pH, and availability of nutrients), an- structures were formed by bacteria of the genus Streptococcus and
timicrobial peptides, and host factors in saliva and gingival crevice sometimes by those of the genus Porphyromonas. Bacteria of the
fluid, including lysozyme, secreted antibodies, and other immune Pasturellaceae family were observed to decorate the apical tips of
|
30       DURAN-PINEDO

A F I G U R E 1   Corncob structures form


around Corynebacterium but do not form
around nearby Fusobacterium, Leptotrichia,
or Capnocytophaga. (A–C) Methacrylate-
embedded sections from 3 different
donors. Rectangles indicate the location
of the insets; ovals and circles are drawn
around representative corncob structures,
each of which has a Corynebacterium core.
Originally published by Mark Welch et al.
B

the filaments from some Corynebacterium species in the "corncob" Treponema are increased, and the relative abundance of bacteria of
structure (Figure 1). Visualization of subgingival plaque represents the genera Dialister, Eubacterium, Selenomonas, and Parvimonas is
an additional challenge. Maintaining the integrity of the structures decreased.35,56 In supragingival plaque, the relative abundance of
in subgingival plaque has been achieved by using carriers in the facultative anaerobic bacteria, of genera including Streptococcus,
dental pocket, thus permitting imaging of the biofilm at different Capnocytophaga, Neisseria, Haemophilus, Leptotrichia, Actinomyces,
developmental stages; the spatial arrangement of the microbiota in Rothia, Corynebacterium, and Kingella, is significantly increased.
periodontal pockets was visualized using electron microscopy, im- A significant fraction of bacteria present in the oral cavity has
munohistochemistry, and fluorescence in situ hybridization.57 These not yet been cultured in the laboratory.63 Despite intensive efforts
findings showed that spirochetes colonize deep within the periodon- by the community of microbiologists studying the oral microbiome,
tal pocket and streptococci colonize at sites closer to the surface. one-third of oral bacterial taxa are as yet uncultivated in vitro.64 The
The initial colonizers were found at the surface of the probe and unculturable condition of a specific bacterium indicates simply that,
later colonizers showed a patchy distribution.58,59 Noiri et al, on their using our current knowledge, we are unable to grow it under labora-
series of microbial localization within periodontal pockets, found tory conditions. Thus, “unculturable” does not indicate the inability
that bacteria were distributed in different regions: Campylobacter of particular organisms to grow in laboratory conditions but rather
rectus was present deep in periodontal pockets; Treponemes reflects our current lack of knowledge of their biology during natural
and Fusobacterium spp. were present in unattached plaque; and growth.65 This “unculturability” could be responsible for underesti-
Actinomyces viscosus and Eikenella corrodens were found on plaque mation of the number of known species. As an example, a fraction
attached to the tooth.60,61 Porphyromonas gingivalis was directly at- of unculturable cells has been reported in 2 easily culturable spe-
tached to the biofilm.62 In supra- and subgingival biofilm, at the genus cies, Fusobacterium nucleatum and Porphyromonas endodontalis. This
level, Corynebacterium and Kingella represent a substantial fraction finding suggests that some of those species included unculturable
of the microbiome, while members of the genus Granulicatella are biotypes, implying that the abundance of species already described
absent in those sites and present in other oral structures.36 could be higher. That a significat fraction of unculturable taxa are
In subgingival sites, the proportions of obligate anaerobic bac- present in gingivitis,66 as well as in periodontitis67,68 has now been
teria of the genera Fusobacterium, Prevotella, Porphyromonas, and confirmed by molecular analysis.
DURAN-PINEDO |
      31

The Tannerella genus comprises 3 species, 1 of which–Tannerella during periodontal disease. These genes included ones that encode
forsythia–has long been known to be highly associated with chronic cell-division proteins (chromosomal replication initiator protein
periodontitis. The other 2 species of this genus–Tannerella spp. DnaA, cell division protein FtsA, and ATP-dependent protease FtsH)
BU045 (human oral taxon 808) and Tannerella spp. BU063 (human and chaperones (GroEL, GroES, DnaJ, and Hsp20). In addition, a
oral taxon 286)–are associated with oral health.9,69 All three spe- number of putative virulence-related genes were also identified that
cies need external factors (provided either by helper species such encode the following: oligopeptide ABC transporter; periplasmic oli-
as Propionibacterium acnes and Prevotella intermedia or by purified gopeptide-binding protein, which has been identified as a virulence
forms of N-acetylmuramic acid on enriched growth medium) for factor in group A streptococci;80 a hemagglutinin-related protein;
growth.64,70,71 As a result of intensive work and use of single-cell exoproteins involved in heme utilization or adhesion; a homolog of
genome sequencing techniques, Tannerella spp. BU045 and BU063, LemA; a widely conserved 2-component regulatory system that has
classically unculturable species, have also been culturable and been described as regulating virulence factors and toxin production
sequenced.71 in Pseudomonas syringae;73 as well as proteins involved in iron acqui-
According to gene content and average nucleotide identity, sition (hemagglutinin-related proteins and an exoprotein involved in
Tannerella phylotypes BU045 and BU063 are more closely related. heme utilization of adhesion) and biosynthesis of pili (PilB and PilC).
However, 2 independent data sets of association with periodon- Also, in periodontitis samples, Saccharibacteria upregulated genes
titis, one based on 16S ribosomal RNA gene abundance and the encoding a chitinase and a putative ROK-family transcriptional reg-
other based on metatranscriptomic data, showed that Tannerella ulator, which regulates chitinase expression in the gram-positive
BU045 is more highly associated with disease than Tannerella pathogen Listeria monocytogenes at the post-transcriptional level.81
64,71
BU063. In contrast to what was believed, these findings sug- These findings suggest a contribution of Saccharibacteria to peri-
gest that Tannerella BU045 is more prevalent in periodontitis than odontal disease; however, the question of whether Saccharibacteria
in oral health. 63,64,71 Interestingly, Tannerella BU063 lacks the vir- have a role in the progression of periodontitis remains unanswered.
ulence genes present in T forsythia, including karilysin, prtH, and Finally, Duran-Pinedo et al75 also observed the over-representation
64,70,71
bspA, which could explain its lack of pathogenicity. of genes involved in pili biosynthesis. Although type IV pili may fa-
Among the 30% of unculturable oral microbial species (http:// cilitate the adherence of bacteria to epithelial cells, it has been sug-
www.homd.Org) are members of the Saccharibacteria phylum gested that in the case of Saccharibacteria, they may be involved
(formerly known as TM7). Recent studies using 16S ribosomal in gliding motility.74 These results highlight the usefulness of in situ
RNA have identified Saccharibacteria in environmental samples metatranscriptomic studies in allowing us to acquire knowledge on
and from eukaryotic cells, including those from the gastrointes- physiology and on structures, such as pili, in the absence of culti-
tinal tract, skin, oral mucosa during active infection, and female vation. By combining the power of single-cell genome sequencing
72
genital tract. Within the oral cavity, at least 6 distinct groups of and metatranscriptomic analysis, we will be able to unveil the role of
Saccharibacteria have been described across anatomic sites.73,74 uncultured bacteria and open the way to a new hypothesis.
The use of new technologies is unveiling the potential role of
Saccharibacteria in periodontal disease. Saccharibacteria spp.
have been found both in healthy sites and in sites with severe peri- 2.1 | Pipeline of metatranscriptome analysis
odontitis.75 One study reports the presence of a high number of
oral Saccharibacteria76 , contradicting previous studies that show The metatranscriptome identifies the gene-expression profiles of
very low abundance.77,78 complex microbial communities based on the set of transcripts being
Recently, a first approach to the unculturable of Saccharibacteria synthesized under diverse growing conditions or environments, in
was reported.79 Despite multiple efforts, the routine unculturable contrast to the metagenome, which only reveals the microbial phy-
of Saccharibacteria remains arduous and challenging. Nanosynbacter logenetic composition of the community (Figure 2). One of the main
lyticus type strain TM7x, a member of the Saccharibacteria phylum, requirements for analysis of the metatranscriptome is availability
has been described as an epibiont of Actinomyces odontolyticus strain of complete genomes of the community to be studied. Fortunately,
(XH001) with a parasitic phase. Depending on environmental con- the oral microbial community is one of the best characterized in the
ditions, TM7x can kill the host bacterium, a feature that may have human microbiome, which makes this type of analysis possible.
an impact on the ecology of the oral microbiome. It is an extremely The first big challenge faced when measuring microbial gene
small coccus (200-300 nm). Its genome (705 kb), the first of a hu- expression in vivo in oral samples is the low biomass. This problem
man-associated Saccharibacteria phylotype to be sequenced, com- can be solved by 2 approaches: the first suggests pooling sam-
pletely lacks the genes required to synthesize amino acids, vitamins, ples with similar clinical profiles to obtain the concentration of
nucleotides, and cell-wall precursors, and therefore it relies on ex- RNA required for the analysis 82; and the second involves linear
79
ternal sources to supply these factors. RNA-amplification methods, which have been successfully used in
Despite the fact that the genomic sequence of candidate division deep sequencing, providing high-quality coverage almost identi-
Saccharibacteria was still incomplete, in 2014, Duran-Pinedo et al75 cal to that of nonamplified libraries. 83 In many cases, an ampli-
were able to identify genes of TM7 that were differentially expressed fication step is required in order to generate sufficient RNA for
|
32       DURAN-PINEDO

F I G U R E 2   Laboratory workflow for


metatranscriptome analyses of microbial
communities. The figure summarizes the
different processes that can be performed
to study the metatranscriptome of the
oral microbiome. mRNA, messenger
RNA; NGS, next-generation sequencing
techniques; rRNA, ribosomal RNA.
Originally published by Solbiati and Frias-
Lopez. 28

F I G U R E 3   Summary of bioinformatic
workflow for metratranscriptomic
analysis. Common steps in the
bioinformatic analysis of microbial
metatranscriptomes start with quality
control of the sequences, alignment of
these sequences against the genomes of
the microbial community, phylogenetic
assignment of the transcripts,
differential expression analysis, and
pathway enrichment analysis. The
software packages widely used for
metatranscriptomic analysis are shown
in blue. HOMD, Human Oral Microbiome
Database; HMO, Human Microbiome
Project; LefSe, linear discriminant analysis
effect size; rRNA, ribosomal RNA.
Originally published by Solbiati and Frias-
Lopez. 28

next-generation sequencing processes. The amplified RNA is a di- Bioinformatic analyses of the metatranscriptome allow large
rect representation of the relative amount of mRNA found in the data sets to be processed through specific steps, called workflow
original clinical sample. 84 or pipeline (Figure 3). As is essential to work only with sequences
Although the application of metatranscriptomics to characterize of optimal quality, the following procedures are undertaken: first,
the functions of microbial communities is increasing rapidly, there is low-quality sequences are removed; second, sequences are aligned
still no consensus on the best approach to perform this type of study. against the genomes of the microbiome being studied; third, a differ-
The approach will depend on the amount and quality of mRNA, the ential expression analysis is performed to identify changes in physio-
success in preparation of the libraries, and the bioinformatic pipeline logic activities; and, fourth, a phylogenetic analysis of the transcripts
utilized in the analyses. is carried out to identify active members of the community.
DURAN-PINEDO |
      33

Along with the bioinformatic analyses of the mRNA sequences, on the expression profile of genes in the community by shotgun
sample size estimation with statistical power is crucial for optimi- metagenomic and metatranscriptomic analyses.36,91 Interestingly,
zation of a metatranscriptomic experiment. In this case, we should we can assess the expression of any gene in a specific species and
consider power wanted, sequencing depth, the coefficient of vari- also determine the functional activities expressed by the whole com-
ation, single- or paired-end reads, effect size, and budget con- munity, acting as a meta-organism. As stated before, not only does
85,86
siderations. The statistical power can be enhanced by using the oral environment or niche shape the diversity of the microbiome,
paired-end reads, and by increasing the sample size and the se- it also modulates the biological pathways within the microbiome.
quencing depth.86,87 To increase the power, the increment in sample For instance, gamma-aminobutyric acid biosynthesis is increased on
size is more potent than sequencing depth, especially when the se- the tongue dorsum. Gamma-aminobutyric acid is a suppressor neu-
86
quencing depth reaches 20 million reads. Unfortunately, in many rotransmitter known to counterbalance the action of the excitatory
clinical trials it is not possible to obtain a large number of replicates. neurotransmitter glutamate. To date, the role of gamma-aminobu-
Nonetheless, the statistical power of deep sequencing provides a tyric acid produced in the oral cavity is still unknown. The supragin-
robust output that can provide highly informative, statistically signif- gival plaque is also enriched in metabolic functions associated with
icant data. Interestingly, in model organisms, statistical power can be environmental information processing (eg, secretion systems, signal
reached with a fairly small number of replicates. For instance, Ching transduction, and molecules involved in signaling) interaction), and
et al compared 5 differential expression analysis packages, evaluat- energy metabolism (eg, oxidative phosphorylation, methane metab-
ing their efficiency using power and other metrics; they found that olism, and nitrogen and sulfur metabolism).
after achieving a sequencing depth of 20 million paired-end reads, all To have a baseline with which the metatranscriptome can be
packages reached a power of > 0.8 with only 10 replicates per con- compared is crucial in order to characterize the transcriptome pro-
dition.86 Using a similar analysis, Heart et al found that the biological file under healthy conditions. Interest in the expression profile of
coefficient of variation of the data sets was a significant driver in health-associated genes is increasing; however, the number of stud-
calculating sample size, but with low coefficients of variation, the ies on patterns of gene expression in vivo remains limited. Benitez-
number of samples per group was also small. With a coefficient of Páez et al92 published 1 of the few studies on characterization of the
variation of 0.4, only 10 samples per condition and 10 million se- metatranscriptome during plaque formation.
quences were needed to reach a power of 0.8. However, with a co- Interestingly, the biofilm-formation process is also determined
efficient of variation of 1.2, 40 samples per condition were needed by person-specific factors after digestion of food, as determined in
to achieve the same statistical power.85 a recent metatranscriptomic analysis.92 In some people, more than
80% of active bacteria belonged to only 3 genera (Actinomyces,
Corynebacterium, and Rothia), whereas in others no predominant
3 |  M I C RO B I A L M E TATR A N S C R I P TO M E I N genera were detected in their active microbial community.
H E A LTH In some individuals, there were virtually no changes in the active
bacterial population after ingestion of food, suggesting that their mi-
The oral cavity is the first contact of the digestive tract with the en- crobiota is not affected by food ingestion, potentially reducing the
vironment and is exposed to countless numbers of microbes through risk of acidic pH and promoting dental health. Benitez-Páez et al92
breathing, eating, and drinking. The commensal oral microbiota may also showed that expression of genes linked to translation machin-
play a role in directing the foreign microbes ingested to flushing sa- ery is higher in early stages of biofilm formation, whereas in mature
liva, and once in the stomach, the extremely low pH will do its part.88 biofilm, genes involved in competence, quorum sensing, mutacin
The commensal microbiome may also modulate colonization of production, and DNA uptake were over-expressed, indicating more
the oral epithelium by extraoral microbes and pathogens through sophisticated levels of interactions in mature biofilm than during
"colonization resistance" or "bacterial interference" processes.89 earlier stages of biofilm formation.
Diverse mechanisms, including competition for nutrients, could also In the majority of cases, active members of the microbial
be responsible for colonization resistance through modulating the community were from the genera Streptococcus (12%-19%) and
microenvironment to inhibit potential competitors and by secretion Actinomyces (3%-12%). Bacteria from the genera Actinomyces
of antimicrobials against these species.89 A significant function of were present at higher frequencies in early plaque samples, con-
the commensal microbiome is to stimulate development of the im- sistent with its known role as an early colonizer. Other phyla from
mune system to maintain a controlled balance between pro- and which active members of the microbial community were fre-
anti-inflammatory responses to bacteria in the absence of active in- quently identified were Actinobacteria (Rothia, Angustibacter, and
fection.90 As a response to colonization resistance, pathogenic bac- Kineococcus) Proteobacteria (Neisseria, Kingella, and Alysiella), the
teria release their arsenal of virulence factors. Firmicutes (Gemella, Paenibacillus, and Veillonella), Bacteroidetes
Currently, because of next-generation sequencing approaches (Capnocytophaga), and Fusobacteria (Fusobacterium).93 In the early
and the availability of genomes for oral microbes, we are able to as- stages of supragingival plaque development, genes involved in the
sess the functional role of the oral microbiome in different environ- metabolism of carbohydrates, energy, amino acids, cofactor/vita-
ments and conditions. Nowadays, we are able to obtain information mins, and xenobiotic degradation were predominantly upregulated.
|
34       DURAN-PINEDO

Moreover, in the late stages of supragingival plaque formation, the transitional state during progression from gingivitis to periodonti-
upregulation of genes involved in the quorum sensing response, in tis. Interestingly, during gingivitis, the oral microbiota expresses not
particular genes identified as belonging to type II secretion systems, only nonspecific peptidases, proteases, and stress response proteins
was reported. As type II secretion systems promote the secretion of associated with colonization, but also putative virulence factors,
folded periplasmic proteins that typically play a role in survival, this such as collagenases, gingipains, and hemagglutinins.82 Virulence-
finding indicates that the community is adapting as it develops. related genes of Leptotrichia buccalis were upregulated, including an-
The influence of oral microbes goes beyond the oral cavity. tibiotic resistance and nonspecific proteases, as well as the response
There is a strong association between the oral microbiome and sys- regulator, MprA (which is strongly associated with pathogenesis in
temic health. The oral microbiome has been implicated in the main- Mycobacterium tuberculosis, suggesting that it may also be involved
tenance of vascular health through the reduction of nitrate to nitric in L buccalis pathogenicity).101 During gingivitis, the expression of
94-96
oxide. Saliva contains high levels of dietary nitrate that may be genes involved in antibiotic resistance, proteolysis, breakdown of
reduced by oral nitrate-reducing bacteria.97 The contribution of the collagen, and iron intake were increased in both F nucleatum and
oral microbiome to the reduction of nitrate remains to be explored.98 Prevotella nigrescens. Prevotella nigrescens also expressed endothe-
We are just at the dawn of functional studies of the oral micro- lin-converting enzyme 1 and a gingipain, and several arginine/ly-
biome. In the years to come, we will have an excellent understand- sine-specific proteases. The expression of those enzymes, which are
ing of how biological activities maintain a healthy microbiome, and related to virulence, could moderate the immune response as they
we are likely to obtain a clearer picture of the transition to disease. are able to degrade host cytokines.102,103
Changes in microbial diversity and gene-expression profiles result- Nowicki et al also described high homology of the expression
ing from that transition could be used as microbial biomarkers for of collagenase with one of the Xaa-Pro aminopeptidases on the
oral infectious diseases. T forsythia genome (BFOR_2955).33 Expression of this protein could
link T forsythia to the initial steps of collagen damage present in
periodontitis.
4 |   M I C RO B I A L M E TATR A N S C R I P TO M E There is no doubt that our current knowledge of the communi-
I N PRO G R E S S I O N O F G I N G I V ITI S A N D ty-level shifts occurring during the transition from health to gingivi-
PE R I O D O NTITI S tis, and later to periodontitis, has increased considerably in the last
decades. However, to unveil the exact process of transition to dis-
Gingivitis is a reversible stage of periodontal disease affecting supra- ease, further exploration in longitudinal studies is needed, involving
and subgingival sulci. Like periodontitis, gingivitis is characterized by a larger number of samples. This exploration should include analysis
inflammation induced by the oral microbiota. However, unlike perio- of unpooled samples, thus allowing an independent study of each
dontitis, gingivitis is initiated at the apical gingival border, namely the site to be performed.
upper limit between the supra- and subgingival areas.90 Classically, The question of why some untreated teeth with clinical manifes-
gingivitis has been correlated with a reduction in, or absence of, oral tation progress to periodontal disease and others remain stable is
hygiene for a short period of time and resolves typically after reini- still unanswered.104 To address this question, research has been fo-
99,100
tiation of a dental prophylaxis routine. New studies have re- cused on identifying markers that might distinguish between active
vealed a stronger correlation between microbial abundance and the and nonactive sites. Those biomarkers include protein activities,105
clinical severity of gingivitis than the oral hygiene status, suggesting bacterial profiles,106,107 cytokine levels,108 genetic markers,109,110
that brushing affects the relative abundance of microbes more than and clinical markers.107 Clinically, the progression of periodontal
33
the diversity of different taxa. The oral biofilm established during disease follows cycles of periods of exacerbation (activity) and re-
gingivitis, can lead to the development of periodontitis. mission.104,111-114 Some studies have suggested that changes in the
Molecular analyses of dental plaque communities in volunteers composition of subgingival biofilm could explain the transition to
with experimentally induced gingivitis have revealed a unique co- disease. However, this hypothesis is invalid because of considerable
hort of microbes exclusively associated with gingivitis, which include overlap in the composition of the microbial communities in progress-
Fusobacterium nucleatum spp. polymorphum, Lachnospiraceae spp., ing and nonprogressing sites.
Lautropia spp., and Prevotella oulorum.100 Recently, Nowicki et al also To elucidate the biological processes linked to the progres-
found that the genera Oribacterium, Leptotrichia, Tannerella, and sion of periodontal disease Yost et al applied metatranscriptomic
Lachnoanaerobaculum are significantly more abundant during gingi- approaches.32 The authors assessed gene-expression profiles in
33
vitis. This finding reveals involvement of new players during the in- plaque samples isolated from the teeth of patients in whom clinical
duction of dysbiosis of the oral microbiome, which occurs at the very progression of periodontal disease was observed. That longitudinal
early transition to periodontal disease. Nowicki et al also focused on metatranscriptomic study was able to provide us with a snapshot on
33
the expression of virulence factors by those species. long-term healthy subgingival sites, unveiling the changes leading to
Metatranscriptomic analysis of gingivitis samples revealed a the progression of periodontitis. Interestingly, sites that remained
mixed picture of expression profiles of specific genes and virulence healthy between the first visit and the time when subsequent
factors in both health and disease, suggesting the existence of a samples were taken showed an unaltered gene-expression profile.
DURAN-PINEDO |
      35

Changes in expression were observed on sites that progressed to important roles in microbial virulence and infection.125,126 Small non-
disease. coding RNAs repress translation of mRNA by attaching to the ribo-
It is noteworthy that differences in the gene-expression profiles some-binding site, competing with the ribosome and leading to the
of baseline (or initial samples) of progressing and nonprogressing rapid degradation of the mRNA.127 Other noncanonical mechanisms
sites were found. In the baseline of progressing sites, Gene Ontology of translation repression have been described, such as the binding
Term Enrichment analysis showed overrepresentation of functions of cis-acting antisense RNA to a ribosome standby site upstream of
related to iron, potassium, chloride, citrate, amino acid transport, the ribosome-binding site.128 Besides, small noncoding RNAs can
lipid A and peptidoglycan biosynthesis, cell motility, and protein activate the translation of mRNAs, or can modulate gene expression
kinase C-activating and G protein-coupled receptor signaling path- by varying the level of transcript stability.129 Small noncoding RNAs
ways. By contrast, in baseline samples from nonprogressing lesions, can also modulate protein activity by mimicking the structures of
the gene ontology terms that were overrepresented were related other nucleic acids and sequestering proteins that otherwise will act
to metal ion transport, the phosphoenolpyruvate-dependent sugar on their real target.130 Diverse experimental approaches have been
phosphotransferase system, tricarboxylic acid, and protein secre- applied to identify bacterial small noncoding RNAs.131,132 Those ap-
32
tion. Interestingly, Yost et al described overexpression of histidine proaches include direct labeling of RNA, and sequencing or collect-
biosynthesis in baseline samples of progressing lesions,, while Jorth ing small noncoding RNA genes by shotgun cloning of their cDNA
et al82 found upregulation of histidine catabolism in disease sites. It and use of microarrays and next-generation sequencing technolo-
is tempting to think that the availability of histidine might represent gies. However, those methods are laborious and expensive, espe-
a signal to disease. cially if a large number of elements are analyzed. Global detection
As a general picture, during health, genes involved in ascorbate of small noncoding RNAs has been facilitated by the development
and aldarate metabolism,115,116 vitamin B6 metabolism and glycoly- of microarrays and next-generation sequencing techniques. Next-
sis, and gluconeogenesis, as well as the pentose phosphate pathway, generation sequencing has allowed the discovery of numerous
pyruvate metabolism, and propanoate and butanoate metabo- novel small noncoding RNA transcripts in different bacteria, such as
lism,82,117,118 were upregulated by the oral microbiota. Salmonella spp.,133 Vibrio cholerae,134 and Bacillus subtilis.135
On sites that progressed to disease, the known periodontal Our knowledge of the small noncoding RNA metatranscriptome
pathogens Treponema denticola and T forsythia were not significantly in complex microbial communities is even more limited than our un-
active, while P gingivalis showed increased activity, supporting the derstanding of the role of small noncoding RNAs produced by spe-
keystone-pathogen hypothesis.32 According to the keystone-patho- cific bacteria. Community-wide studies have been mainly limited
gen hypothesis, specific low-abundance bacteria can transform an to controlled environments.136,137 The first community-wide meta-
ordinarily healthy microbiome into a dysbiotic state, causing peri- transcriptomic analysis of small noncoding RNAs by Shi et al136 was
32,119
odontal inflammation and tissue destruction. The increased reported in samples from the ocean. In that study, the researchers
activity of P gingivalis in initial samples of active sites supports the found a large number of previously unknown, putative small non-
hypothesis that P gingivalis leads to the dysbiotic process which initi- coding RNAs, as appropriate. Gosalbes et al reported the presence
ates the disease.120,121 It is tempting to think that particular proteins of small noncoding RNAs in the metatranscriptome of human fecal
expressed by P gingivalis could be a bacterial biomarker for periodon- microbiota, although these researchers did not identify the possible
titis. In this regard, more extensive longitudinal studies are neces- functions of the small noncoding RNAs.138 Research on small non-
sary to identify the expression of hallmark genes that will allow us coding RNAs in the oral microbiome is very limited. Based on their
to track the progression of periodontal disease at a particular site. study of expression profiles in periodontitis and in health, Duran-
A less studied aspect of the survival and physiological adapta- Pinedo et al identified small noncoding RNAs by mapping transcripts
tion of microorganisms in the oral cavity is the regulatory functions to an Intergenic region (IGR) database generated from the genomes
of bacterial small noncoding RNAs. Those elements are rapidly syn- used in that analysis.84 The authors reported a high correlation be-
thesized and capable of modulating the microbial metabolism faster tween expression profiles of mRNAs and several clusters of small
than the 2-component systems or any regulatory mechanisms for noncoding RNAs. Moreover, the functions of those clusters were
which protein synthesis is required.122,123 Small noncoding RNAs surprisingly similar to signature functions described on the mRNA
comprise a diverse group of RNA molecules with high heterogene- metatranscriptome during periodontitis progression,32 highlighting
ity in size and structure, which do not become translated to pro- the hypothesis that the differentially expressed small noncoding
teins. This is especially crucial in the oral cavity, where conditions RNAs identified participate in the regulation of functions. The small
change constantly, and over short periods of time, because of in- noncoding RNA clusters found were associated with ferrous iron
gestion of nutrients, talking, and the fact that the oral cavity is an transport, proteolysis, potassium ion transport, beta-lactam catab-
open entrance in contact with the external environment. It is now olism, cell adhesion, protein secretion and import, and cobalamin
well established that small noncoding RNAs could regulate physio- biosynthesis, activities that have all been identified in progressing
logical roles, including adaptation to new environmental conditions and severe periodontitis.84
124
through quorum-sensing systems or the development of biofilms. Additionally, gene ontology terms associated with pathogene-
Some reports have shown that small noncoding RNAs also play sis were reported, supporting the hypothesis that small noncoding
|
36       DURAN-PINEDO

RNAs play a key role in the regulation of bacterial activity during Duran-Pinedo et al31 reported an increase in metabolic activities
periodontitis progression. The association of small noncoding RNAs related to iron acquisition, lipopolysaccharide synthesis, flagella
with the regulation of bacterial virulence has been described for in- synthesis, and expression of genes related to beta-lactam antibi-
dividual species such as Staphylococcus aureus,139-141 Pseudomonas otic degradation processes, as the significant activities defining se-
aeruginosa,142 and V cholerae.134,143 vere periodontitis. Iron is an essential enzyme cofactor and many
Small noncoding RNAs have been associated with the regulation microbial pathogens have developed a rage of strategies to acquire
of conjugative transposons in Bacteroides spp.144 High levels of ex- it.154,155 The presence of high levels of bacterial lipopolysaccharide
pression of transposases have been reported in F nucleatum and T has been described as an essential element in periodontitis.156,157 In
denticola145,146; furthermore, in P gingivalis, small noncoding RNAs P gingivalis, high levels of lipopolysaccharide and lipid A have been
147
have been found within putative transposons. described as a mechanism to modulate the inflammation in peri-
These results suggest that small noncoding RNAs might play a odontal tissues by delaying neutrophil apoptosis.158-160
key role in modulation of the oral microbiome from a commensal Isolation of beta-lactamase-producing bacteria from the oral
state to a dysbiotic one by controlling survival, response to the host cavity has previously been reported.161 Moreover, beta-lactamase
immune system, and virulence activities. activity has been observed at a low level, but with high prevalence,
in patients with periodontitis.162 This raises the question of the
role of beta-lactamase in periodontitis as none of the patients was
5 |   M I C RO B I A L M E TATR A N S C R I P TO M E I N being treated with beta-lactam antibiotics at the time of sampling.
PE R I O D O NTITI S Duran-Pinedo et al31 also analyzed the expression of virulence fac-
tors during periodontal disease, and demonstrated upregulation of
Periodontitis is a progressive and chronic disease induced by the metalloproteases and peptidases in bacteria of the red complex.
overgrowth of oral microbiota within the gingival sulcus, resulting Red complex bacteria also expressed proteins involved in iron me-
in the formation of an inflammatory infiltrate that leads to the de- tabolism, including ferrous iron transport protein B, iron compound
struction of the periodontium and alveolar bone, and may eventu- ABC transporter ATP-binding proteins, ferric enterobactin transport
ally result in tooth loss.148,149 During this process, oral homeostasis ATP-binding protein FepC, and iron chelate uptake ABC transporter,
is disrupted, and the local microbiota changes from symbiosis to a FeCT family, permease protein. Porphyromonas gingivalis upregu-
dysbiotic state, leading to disease. The transition of the oral micro- lated expression of hemolysin genes, and P gingivalis and T denticola
biome from commensal to pathogenic is a dynamic process between showed upregulated expression of genes involved in vitamin B12
the complete oral endogenous microbiota and the host response.119 synthesis (for example, vitamin B12 ABC transporter btuFCD sys-
Classically, periodontal disease has been associated with bac- tem). Among the proteins not related directly to iron metabolism
teria of the so-called red complex (P gingivalis, T forsythia, and were ATP-dependent Clp protease ATP-binding subunit ClpA and
T denticola). Currently, these microorganisms are known as patho- chaperone protein ClpB, described as necessary in cellular adhesion
bionts rather than pathogens because they are present at low and invasion of P gingivalis. Treponema denticola overexpressed genes
numbers in microbiomes of patients with no clinical signs of peri- related to flagella synthesis, such as flgC, flgF, and flgG, the flagellar
150
odontal disease. The pathobionts group now includes anaerobes hook proteins FlgE and FlgK, the flagellar motor rotation proteins
of the genera Parvimonas, Fusobacterium, and Prevotella as well as MotA and MotB, the flagellar motor switch proteins FliG and FliM,
gram-positive bacteria such as Filifacter alocis.35,148,151 Periodontitis flagellin protein FlaA, flagellar M-ring protein FliF, flagellum-specific
is characterized by changes in the structure of the microbial commu- ATP synthase FliI, and proteins involved in flagellum biosynthesis,
nity; namely an increase in the number of bacterial taxons and in the supporting the advance of periodontal disease by continued inva-
relative abundance of pathobionts.148,152,153 As in other infectious sion of the tissue. Tannerella forsythia and T denticola were found
diseases, the microbial diversity has been studied in detail, in con- to upregulate homologs to internalins, surface proteins that are in-
trast to the bacterial gene-expression profiles in vivo, which remain volved in the invasion of mammalian cells by L monocytogenes163,164
less explored. Fortunately, the application of metatranscriptomics to and which could play a similar role in those periodontal pathogens.
study the functional activities associated with periodontal disease These observations strongly support the idea that in severe peri-
has increased in the last few years. Despite high interpatient vari- odontitis, members of the ‘red complex' actively invade host cells.
ability in the oral microbiome, those studies suggest that we should There is no doubt that the most intriguing result of that work was
focus more on the functional changes of the whole community than the upregulation of a vast majority of virulence factors by organ-
on specific pathogens or the microbial composition.32,75,82 isms not considered as pathogens. Surprisingly, the gram-positive
Recently, the phylogeny and diversity of the oral microbiota have bacteria Corynebacterium matruchotii, Rothia dentocariosa, Veillonella
been characterized at unprecedented levels using high-throughput parvula, and Neisseria sicca, classically associated with the health of
sequencing. Early metabolic activities in the dysbiotic microbiome the periodontium, expressed a large variety of putative virulence
revealed a functional signature that distinguishes periodontal sites factors. Interestingly, candidate division TM7 (Saccharibacteria), not
from healthy ones, supporting the idea that microbial commu- cultivated at the time of that publication and with only a partially
nities as a whole might drive disease progression.75 In that study, sequenced genome, showed overexpression of several homologs
DURAN-PINEDO |
      37

to virulence factors in the diseased community.31 These included: during periodontal disease progression. Specifically, genes with gene
the oligopeptide ABC transporter periplasmic oligopeptide-bind- ontology terms associated with transport (iron, lactate, citrate, so-
ing protein, previously described as a virulence factor in group A dium, and phosphate), proteolysis, the protein kinase C-activating
Streptococci165; a hemagglutinin-related protein; exoproteins in- G-protein coupled receptor signaling pathway, and response to
volved in heme utilization or adhesion; and a homolog of LemA, a antibiotic were upregulated; and genes with gene ontology terms
2-component regulatory system identified as responsible for regu- associated with cobalamin (vitamin B12) biosynthesis were down-
lating virulence factors and toxin production in Pseudomonas syrin- regulated. Motility of T denticola was indicated by the upregulation
gae.73 Those results support the proposed idea of the “pathogenic of genes related to flagella biosynthesis (flaN, flaG, fliQ, and fliW).
microbial community”166 or “the community as pathogen”,167 which Tannerella forsythia and P gingivalis both upregulated different Ton
postulates that the integrated actions of the components of the mi- B-dependent receptors, genes involved in iron transport (ferric up-
crobial community would result in disease (Figure 4). take siderophores and ferrous iron transport protein B), and a large
Later on, Yost et al32 showed a general picture of the bacterial number of peptidases and proteases, including ClpB. They also upreg-
pathways associated with progression to periodontitis. The authors ulated genes associated with aerotolerance, and clustered regularly
analyzed the metatranscriptome in subgingival samples from sta- interspaced short palindromic repeat-associated genes (csp, csm, and
ble and progressing sites. Disease was considered to be progress- cas).32 Finally, P gingivalis specifically upregulated genes related on
ing if an increase in clinical attachment loss, of ≥2  mm compared biotin synthesis (biotin synthase, bioC, and bioG), proteins involved
with baseline, was recorded at any follow-up visit. Stable sites were in the biosynthesis of capsular polysaccharide, a large number of
defined as demonstrating clinical attachment loss no greater than proteins of conjugative transposons (TraA, TraB, TraE, TraF, TraG,
1 mm from the measuremnet made at baseline. The authors ana- TraI, TraJ, TraK, TraL, TraM, TraN, TraO, Trap, and TraQ), transposases
lyzed the gene-expression profile of the community by identifying (ISPg2, ISPg3, ISPg4, ISPg5, and ISPg6), and the 2 clustered regu-
the gene ontology terms that were enriched. Changes took place in larly interspaced short palindromic repeat-Cas operons (CRISPR-cas)
the gene-expression profiles within all members of the red complex found in P gingivalis ATCC 33277. Porphyromonas gingivalis showed

A B

F I G U R E 4   Species ranked according to the number of upregulated putative virulence factors in the metatranscriptome. Putative
virulence factors were identified by aligning the protein sequences from the different genomes against the Virulence Factors of Pathogenic
Bacteria Database. The numbers in the bar chart are the absolute number of hits, for each bacterial species, of the upregulated putative
virulence factors identified. Members of the red complex are shown in red, members of the orange complex are shown in orange, and
members of the yellow complex are shown in blue. (A) Comparison of health with severe periodontitis (adapted from Duran-Pinedo et al75).
(B) Comparison of baseline with sites showing progression of periodontitis (adapted from Yost et al32).
|
38       DURAN-PINEDO

upregulation of a large number of genes associated with virulence in regularly interspaced short palindromic repeat-associated genes (in
healthy sites that later progressed to periodontitis, while T forsythia Centruroides gracilis, Campylobacter rectus, Campylobacter showae,
and T denticola upregulated only one of a few virulence genes until P nigrescens, and Streptococcus constellatus), and the chaperone pro-
later time points in this study.Individually, T denticola upregulated teins GroEL, GroES, and GrpE. As in the case of P gingivalis, both P in-
genes related to flagella biosynthesis (flaA, flaG, fliQ, and fliW), oligo- termedia and P nigrescens upregulated a large number of mobilization
peptide ABC transporters, and a large number of hypothetical pro- genes (traA, traB, traD, traE, traF, traG, traI, traJ, traK, traL traM, traN,
teins. Tannerella forsythia and P gingivalis both upregulated different traO, and traQ) from conjugative transposons.32
TonB-dependent receptors, genes involved in iron transport (ferric Interestingly, organisms not associated with periodontal diseases,
uptake siderophores and ferrous iron transport protein B), a large such as Streptococcus oralis, Streptococcus mutans, Staphylococcus in-
number of proteases such as ClpB, and clustered regularly inter- termedius, Streptococcus mitis, Veillonella parvula, and Pseudomonas
spaced short palindromic repeat-associated genes (csp, csm, and cas). fluorescens, were particularly active, transcribing genes related to
Tannerella forsythia specifically upregulated transposases (IS116, virulence. All of the above-cited organisms upregulated genes en-
IS110, IS902, and IS4 families) and large numbers of homologs of coding different oligopeptide transport systems (oppA, oppD, oppF,
32
SusC and SusD family proteins involved in polysaccharide binding. and oppB), several hemolysins, manganese ABC transporters, man-
In addition, T forsythia significantly upregulated the expres- ganese superoxide dismutase, and a protein serine-threonine phos-
sion of genes related to the destruction of the periodontium. The phatase (PrpC) involved in regulating the stationary phase of the cell
bfor_c_1_1659 open reading frame was highly expressed in sites that cycle. Streptococcus oralis, S mitis, and V parvula upregulated vitamin
31,32
progressed to periodontitis. BFOR_c_1_1659 has homology to B12 ABC transporters. Streptococcus oralis, S mutans, S intermedius,
dipeptidyl aminopeptidase IV, a serine protease that cleaves X-Pro and S mitis all upregulated the Clp protease system and the tran-
or X-Ala dipeptide at the penultimate position from the N-terminal scription attenuator LytR. Pseudomonas fluorescens upregulated the
ends of polypeptide chains. Collagen, rich in X-Pro or X-Ala dipep- complete set of genes associated with flagellar synthesis (flaA-S and
tide, is the main supporting structure in connective tissues, which motB) and genes related to chemotaxis.32 These findings suggest
168
are present on all structures of the periodontium. The ability that the whole community, not just a handful of oral pathogens, is
of BFOR_c_1_1659 to degrade collagen was recently assessed. responsible for the increase in virulence that leads to progression of
Expression of this putative collagenase was only found in sites with periodontal disease.
169
severe periodontitis. Tannerella forsythia also specifically upregulated several trans-
By contrast, during gingivitis, T forsythia downregulated BFOR- posases (IS116, IS110, IS902, and IS4 families), and large numbers
1659 and upregulated another Xaa-dipeptidase (BFOR-2955), as de- of different homologs of SusC and SusD family proteins that are in-
scribed by Nowicki et al33. Those results suggest that the ability of volved in polysaccharide binding. Regarding proteins downregulated
T forsythia to contribute to the destruction of collagen could be a by red complex bacteria, mostsuch proteins were hypothetical.31
result of selective expression of proteins triggered by still unknown Archaea have not been as widely studied as Bacteria although
mechanisms. The over-representation of gene ontology terms re- they might have a direct effect on the development of periodonti-
lated to potassium ion transport is also an essential signature of tis. Archaea are found in 36% of patients with periodontitis, with
31,32
periodontal disease. Potassium transport systems have been the Methanobrevibacter oralis-like phylotype being the predominant
linked to the pathogenesis of S aureus170 and Salmonella171, but inter- archaeal community at periodontally diseased sites; moreover, a dis-
estingly have not been described in oral bacteria. The link between tinct Methanobrevibacter subpopulation is mainly found in the gut of
potassium transport and bacterial pathogenesis is correlated to the mammals. Methanobrevibacter oralis has been found in periodontal
significant increase in levels of potassium in both saliva and ginival pockets and in dental root canals.174 The two main species of Archaea
172
crevicular fluid (GCF) during periodontitis. It is tempting to hy- identified so far in humans are M oralis and Methanobrevibacter
pothesize that higher levels of potassium could increase the viru- smithii. They can colonize the intestine, oral cavity, and skin, and
lence of the oral community. At the same time, microbial imbalance can favor the growth of fermenting bacteria through interspecies
can alter the immune response of the gingival epithelium, increasing hydrogen transfer. There is very little information on the relationship
the production of tumor necrosis factor-alpha and reducing the ex- between Archaea and periodontitis or on the possible mechanisms
pression of interleukin-6 and the antimicrobial peptide human be- of interaction between Archaea and bacteria.175
ta-defensin 3.173
The gene-expression profiles in bacteria of the orange com-
plex are very similar to those in bacteria of the red complex. Both 6 | TH E M I C RO B I A L
upregulate different TonB-dependent receptors, a large number M E TATR A N S C R I P TO M E I N D E NTA L C A R I E S
of peptidases and proteases (including ClpB), genes associated
with aerotolerance (Bacteroides aerotolerance operon batA-E i and The prevalence of caries in permanent teeth has been calculated to
moxR-like ATPase of the aerotolerance operon in P intermedia and be 34%; thus, caries affects 2.5 billion people worldwide. 20 Caries
P nigrescens), genes involved in iron transport (ferric uptake sidero- is characterized by lesions on tooth enamel and dentin induced by
phores and ferrous iron transport protein B), hemolysins, clustered an sugar rich diet. Saccharolytic bacteria in the oral microbiome
DURAN-PINEDO |
      39

ferment sucrose and other sugars to produce ATP and lactic acid as each sample expressed between 70 and 100 metabolically active
a waste product. The accumulation of lactate is responsible for the species associated with oxidative stress. They reported high expres-
local acidification.176 Lowering the pH (to below 5.5) within the bio- sion of proteins that metabolize superoxides and peroxides present
film177 for extended periods of time favors the growth of acidogenic in enamel/coronal caries.
and acid-tolerant (aciduric) bacteria.178 A number of studies on dental caries have shown the presence of
Historically, only acidogenic species of the genus Streptococcus, many activities associated with oxidative stress and that high levels
mainly S mutans, have been considered as the etiological agents of of proteins which metabolize superoxides and peroxides are pres-
dental caries. Recent studies using metatranscriptomics and metabo- ent in enamel/coronal caries.180,185 The functional profiles of car-
lomics approaches found a much higher diversity of bacteria, than pre- ies-associated bacterial communities indicate that genes involved in
viously considered, in alkali-generating pathways within complex oral production of proteins implicated in oxidative stress and metabolism
biofilms, thus linking caries to a more diverse microbiota. Interestingly, of superoxides and peroxides are over-represented only at the ini-
the composition of microbes in the oral biofilm changes at different tial stage (enamel caries). During later stages of caries, expression
stages of caries development.179,180 In contrast to what is observed of genes coding for osmotic stress tolerance proteins as well as pu-
in periodontal diseases, the initial stage of caries is characterized by tative collagenases, arginine deaminase, and urease counterbalance
higher microbial diversity. Caries progression is accompanied by a loss the acidic pH, enabling degradation of dentin in dentin cavities.168
of diversity and a reduction in the levels and activity of commensal The classical mechanism against acidification of the ecosystem was
bacteria.181,182 Once the caries lesion reaches the dentin, diversity thought to be through the production of ammonia (an alkali) from
may increase again, highlighting the influence of the oral niches on arginine and urea.186 However, Edlund et al found that glutamate de-
the activity of the oral microbiome. For instance, in dentin, the rela- hydrogenase, threonine, and serine deaminase, and upregulation of
tive proportion of S mutans increases from 0.12% in healthy dental membrane proteins involved in ammonia gas conduction, acted as a
plaque to 0.72% in enamel lesions.34,183 By metatranscriptomic anal- way to control pH besides the urease activity and arginine deaminase
34
ysis, Simón-Soro et al identified the metabolically active bacterial system.187 It is becoming clear that caries is a more dynamic process
fraction in caries lesions at different stages of disease progression to than originally thought, being characterized by a shift in composition
reveal a big picture of the bacterial community causing tooth decay. of the supragingival microbial community that induces the caries le-
They analyzed active lesions (identified by their color and texture) sion on enamel. Numerous metagenomic studies support the strong
and active white spot lesions or noncavitated enamel caries (iden- correlation between S mutans and Lactobacillus spp. and active car-
tified based on their chalky white, opaque, and rough texture), on ies.181,182,188 At low concentrations of fermentable carbohydrates,
which the carious lesion might extend into dentin without a clinically S mitis bind, with higher avidity than S mutans, to teeth coated with
visible crack at the enamel surface.184 Their data showed variation saliva. Then, S mitis rapidly increase in number, and by secreting
in bacterial consortia between caries lesions of the same individuals hydrogen peroxide antagonize competitors.119,189 The presence
and among individuals. Lactobacilli were found almost exclusively in of acidogenic and aciduric oral microbes, in conjunction with fre-
cavities in dentin. Streptococci accounted for 40% of the entire active quent consumption of dietary sugars, results in caries lessons.
community in enamel caries and 20% in dentin caries. As anticipated, Recent studies present evidence that other species than S mutans
white spot lesions appeared to be a restrictive environment, whereas and S mitis are likely to play a roles in caries development.188,190,191
open dentin cavities had the most diverse bacterial communities. They include: Actinomyces, Fusobacterium, Porphyromonas, Rothia,
Open dentin lesions also showed the highest variability of bacteria be- Granulicatella, Gemella, Selenomonas, Bifidobacteria, Scardovia, and
cause of their exposure to saliva. Streptococci, Rothia, Leptotrichia, and Haemophilus, well as nonaciduric genera including Corynebacterium
Veillonella, for example, were more abundant in carious enamel lesions, spp, Granulicatella spp, Propionibacterium spp., Bifidobacterium spp.,
whereas Lactobacillus, Shlegelella, Pseudoramibacter, and Atopobium and Scardovia spp. Actinomyces and Candida albicans.190 Actinomyces
were associated with dentin lesions. The minority species that were and Candida albicans are strongly associated with caries progression
only present in enamel lesions included Tannerella, Olsenella, Filifactor, in vivo, especially in the case of early childhood caries.192 Similarly
and Treponema, all of which are strongly associated with periodontal to periodontitis, an extended group of species becomes more
disease. pathogenic.
Interestingly, the level of Streptococcus sanguinis was markedly Supporting those results, Kressirer et al found in 2018, that
increased in dentin cavities, whereas S mitis was more abundant in genes expressed in caries mapped to 108 named species.190 Simón-
enamel lesions. Regarding S mutans (the most studied caries-associ- Soro et al34 also found that Streptococcus and Veillonella were highly
ated species), the authors reported a low prevalence in all samples: active in all types of caries, whereas Lactobacillus spp. were only
0.73% in enamel lesions, 0.48% in open dentin lesions, and 0.02% in highly active in the enamel-dentin caries sites. In a parallel study on
hidden dentin lesions.34 Those data suggest that the etiology of den- caries lesions in children, Kressirer et al190 reported that Lactobacillus
tal caries is a very dynamic process shaped by the tissue, and it has spp. are highly active in coronal caries, while in dentin caries,
a polymicrobial origin. The cariogenic properties of the bacteria re- the most representative members of the active community are
cently identified to be associated with caries should be investigated Neisseriales, Prevotellaceae, and Actinobacteria. Scardovia wiggsiae
in future experimental work. Simón-Soro et al34 also reported that is strongly associated with dentin caries, suggesting a role in caries
|
40       DURAN-PINEDO

development.183 By contrast, in a previous study, when looking at some organisms are highly associated with disease, changes in bio-
community-wide expression profiles of dental plaque, Peterson film composition alone do not explain the onset or development of
et al193 found that a small fraction of the community is responsible disease.
for the majority of transcripts in the community. Streptococci rep- It is now clear that the functions of distinct species within the
resent the majority of the active members in caries. Streptococcus subgingival microbiota are intimately intertwined with the rest of
sanguinis was found to be the most active member of the commu- the microbial community. This point highlights the relevance of ex-
nity, with 16% of the transcripts, followed by S mitis (10%), V parvula amining the gene expression profile of the subgingival microbiota in
(9%), Capnocytophaga spp. (9%), S oralis (8%), Streptococcus spp. (7%), periodontal disease or caries lesions in the context of other mem-
Gemella haemolysans (5%), Streptococcus gordonii (4%), and Neisseria bers of the biofilm in vivo.
185
spp. (3%). Metatranscriptomic analysis of the oral community has allowed
The gene-expression profile of caries lesions reported by us to begin answering questions of how homeostasis is maintained.
Kressirer et al190 showed a higher diversity of genes in dentin than in These analyses are the starting point for identifying environmental
caries-free samples or in caries samples from coronal sites. In den- signals that modulate the metabolic patterns of the community as it
tin lesions, larger number of enzymes were detected. This finding shifts from commensal to dysbiotic. This wealth of information was
is consistent with other observations of acidogenic and proteolytic unimaginable just a decade ago.
activities in dentin caries.92 These studies give a shot of the gene expression patterns of
Caries lesions showed upregulation of genes involved in the microbial communities and also allow us to determine triggers to
metabolism of fructose and mannose, sugar phosphotransferases diseases. In the case of caries, new observations on sugar metabo-
involved in the metabolism of beta-glucoside, as well as the use lism (classically identified as an essential element that distinguishes
of sorbitol as an additional source of carbon in caries lesions. May health from disease), were made, such as the potential use of sorbitol
et al194 confirmed this observation on their analysis of already ex- as an additional source of carbon by S mutans. In the case of peri-
isting metatranscriptomic data. In S mutans, the authors found a odontal disease, high levels of extracellular potassium could be a sig-
strong association of the pathway that converts sorbitol to fructose nal of disease. These are just some examples, among other dysbiotic
6-phosphate with caries lesions. This Kegg Orthology (KO) group signals identified by metatranscriptomic analysis.
converts fructose to fructose 6-phosphate, suggesting that in den- Thus, in the near future, the application of metatranscriptomic
tal caries sorbitol might be used as an additional source of carbon. analysis along with other omics approaches will continue to pro-
They also described 9 KEGG Orthology groups from pathways of the vide the opportunity to identify the group of metabolic pathways
phosphotransferase system and mannose metabolism that were as- that trigger the development of periodontal disease and caries.
sociated with caries lesions. Those genes have also been associated Longitudinal studies are needed to identify the real markers of the
195,196
with the colonization process. initial stages of oral diseases. Of note, the metatranscriptome does
By contrast, in caries-free samples or healthy sites, alcohol dehy- not necessarily represent the final metabolic products generated by
drogenase and methylenetetrahydrofolate reductase genes were sig- the microbial community. Improvements in the integration of omics
nificantly upregulated. Alcohol dehydrogenase has been described approaches should allow us to determine the origin of proteins and
in the neutralization of acid through the reduction of nicotinamide metabolites produced by the microbiome in health and disease.
adenine dinucleotide (NAD+ to NADH). Alcohol dehydrogenase is Finally, we need more information on the gene-expression profiles
mainly produced by N sicca.197 Genes expressing methylenetetrahy- of the host, along with the patterns from the microbiome, to obtain a
drofolate reductase are involved in methionine production and are better view of the modulation of health and disease. Understanding
190
mainly expressed by S sanguinis. the communication between the host and the microbiome is cru-
cial to complete the picture between oral health and disease. Finally,
there is a missing piece on the transcriptome, namely the fungal frac-
7 |  CO N C LU S I O N S tion of the oral microbiome, which could influence the dynamics of
the whole microbial community.
Current concepts of caries and periodontal disease highlight the
importance of oral biofilms as and its participation as etiological AC K N OW L E D G E M E N T S
agents of those diseases. There has been a shift in focus from the I thank Jorge Frias-Lopez and Susan Yost for their helpful com-
role of individual species within the biofilm to a more complex ments on this manuscript. This work was supported by the National
view, in which biofilms as a whole cause and perpetuate these Institute of Dental and Craniofacial Research of the National
chronic infections. Institutes of Health (2R01DE021553).
Despite the relevance of the biofilm to the infectious oral dis-
ease, it is clear that not all compositions of the oral microbiota re- C O N FL I C T S O F I N T E R E S T
sult in tissue destruction, and that not all elements of a pathogenic The author declares no potential conflicts of interest with respect to
biofilm have the same significance in the disease process. Although the authorship and/or publication of this article.
DURAN-PINEDO |
      41

REFERENCES 23. McCarren J, Becker JW, Repeta DJ, et al. Microbial community
transcriptomes reveal microbes and metabolic pathways associ-
1. Sender R, Fuchs S, Milo R. Revised estimates for the num-
ated with dissolved organic matter turnover in the sea. Proc Natl
ber of human and bacteria cells in the body. PLoS Biol.
Acad Sci U S A. 2010;107(38):16420-16427.
2016;14(8):e1002533.
24. Carlisle EM, Poroyko V, Caplan MS, Alverdy J, Morowitz MJ, Liu D.
2. Lederberg J, McCray A. Ome sweet ’omics: – A genealogical trea-
Murine gut microbiota and transcriptome are diet dependent. Ann
sury of words. Scientist. 2001;15:8.
Surg. 2013;257(2):287-294.
3. Wade WG. The oral microbiome in health and disease. Pharmacol
25. Giannoukos G, Ciulla DM, Huang K, et al. Efficient and robust
Res Off J Ital Pharmacol Soc. 2013;69(1):137-143.
RNA-seq process for cultured bacteria and complex community
4. Abeles SR, Robles-Sikisaka R, Ly M, et al. Human oral vi-
transcriptomes. Genome Biol. 2012;13(3):R23.
ruses are personal, persistent and gender-consistent. ISME J.
26. Xiong X, Frank DN, Robertson CE, et al. Generation and analysis
2014;8(9):1753-1767.
of a mouse intestinal metatranscriptome through illumina based
5. Ly M, Abeles SR, Boehm TK, et al. Altered oral viral ecology in as-
RNA-sequencing. PLoS One. 2012;7(4):e36009.
sociation with periodontal disease. MBio. 2014;5(3):e01133-1114.
27. Booijink CCGM, Boekhorst J, Zoetendal EG, Smidt H, Kleerebezem
6. Wade WG. The oral microbiome in health and disease.
M, de Vos WM. Metatranscriptome analysis of the human
Pharmacological Research. 2013;69 (1):137-143. http://dx.doi.
fecal microbiota reveals subject-specific expression profiles,
org/10.1016/j.phrs.2012.11.006
with genes encoding proteins involved in carbohydrate me-
7. Lepp PW, Brinig MM, Ouverney CC, Palm K, Armitage GC, Relman
tabolism being dominantly expressed. Appl Environ Microbiol.
DA. Methanogenic Archaea and human periodontal disease.
2010;76(16):5533-5540.
Proc Natl Acad Sci U S A. 2004;101(16):6176-6181. https://doi.
28. Solbiati J, Frias-Lopez J. Metatranscriptome of the oral microbi-
org/10.1073/pnas.03087​66101
ome in health and disease. J Dent Res. 2018;97(5):492-500.
8. Jenkinson HF. Beyond the oral microbiome. Environ Microbiol.
29. Belstrøm D, Fiehn N-E, Nielsen CH, et al. Differences in bacterial
2011;13(12):3077-3087.
saliva profile between periodontitis patients and a control cohort.
9. Paster BJ, Boches SK, Galvin JL, et al. Bacterial diversity in human
J Clin Periodontol. 2014;41(2):104-112.
subgingival plaque. J Bacteriol. 2001;183(12):3770-3783.
30. Belstrøm D, Fiehn N-E, Nielsen CH, et al. Altered bacterial profiles
10. Peterson J, Garges S, Giovanni M, et al. The NIH human microbi-
in saliva from adults with caries lesions: a case-cohort study. Caries
ome project. Genome Res. 2009;19(12):2317-2323.
Res. 2014;48(5):368-375.
11. Dahlén G. Bacterial infections of the oral mucosa. Periodontol.
31. Duran-Pinedo AE, Chen T, Teles R, et al. Community-wide tran-
2000;2009(49):13-38.
scriptome of the oral microbiome in subjects with and without
12. Dewhirst FE, Chen T, Izard J, et al. The human oral microbiome. J
periodontitis. ISME J. 2014;8(8):1659-1672.
Bacteriol. 2010;192(19):5002-5017.
32. Yost S, Duran-Pinedo AE, Teles R, Krishnan K, Frias-Lopez J.
13. Aas JA, Paster BJ, Stokes LN, Olsen I, Dewhirst FE. Defining
Functional signatures of oral dysbiosis during periodontitis pro-
the normal bacterial flora of the oral cavity. J Clin Microbiol.
gression revealed by microbial metatranscriptome analysis.
2005;43(11):5721-5732.
Genome Med. 2015;7(1):27.
14. Dewhirst FE, Paster BJ, Tzellas N, et al. Characterization of novel
33. Nowicki EM, Shroff R, Singleton JA, et al. Microbiota and meta-
human oral isolates and cloned 16S rDNA sequences that fall in
transcriptome changes accompanying the onset of gingivitis. MBio.
the family Coriobacteriaceae: description of olsenella gen. nov.,
2018;9(2):e00575.
reclassification of Lactobacillus uli as Olsenella uli comb. nov. and
34. Simón-Soro A, Guillen-Navarro M, Mira A. Metatranscriptomics
description of Olsenella profusa sp. nov. Int J Syst Evol Microbiol.
reveals overall active bacterial composition in caries lesions. J Oral
2001;51(Pt 5):1797-1804.
Microbiol. 2014;6:25443.
15. Haffajee AD, Socransky SS. Introduction to microbial aspects of
35. Griffen AL, Beall CJ, Campbell JH, et al. Distinct and complex bac-
periodontal biofilm communities, development and treatment.
terial profiles in human periodontitis and health revealed by 16S
Periodontol. 2000;2006(42):7-12.
pyrosequencing. ISME J. 2012;6(6):1176-1185.
16. Haffajee AD, Socransky SS, Patel MR, Song X. Microbial
36. Segata N, Haake SK, Mannon P, et al. Composition of the adult di-
complexes in supragingival plaque. Oral Microbiol Immunol.
gestive tract bacterial microbiome based on seven mouth surfaces,
2008;23(3):196-205.
tonsils, throat and stool samples. Genome Biol. 2012;13(6):R42.
17. Paster BJ, Olsen I, Aas JA, Dewhirst FE. The breadth of bacterial
37. Zhou Y, Gao H, Mihindukulasuriya KA, et al. Biogeography
diversity in the human periodontal pocket and other oral sites.
of the ecosystems of the healthy human body. Genome Biol.
Periodontol. 2000;2006(42):80-87.
2013;14(1):R1.
18. Socransky SS, Smith C, Martin L, Paster BJ, Dewhirst FE, Levin
38. Li K, Bihan M, Yooseph S, Methé BA. Analyses of the microbial di-
AE. Checkerboard DNA-DNA hybridization. Biotechniques.
versity across the human microbiome. PLoS One. 2012;7(6):e32118.
1994;17(4):788-792.
39. Smith QT, Geegan SJ. Repeated measurement of crevicular fluid
19. Eke PI, Thornton-Evans GO, Wei L, Borgnakke WS, Dye BA, Genco RJ.
parameters at different sites. J Clin Periodontol. 1991;18(3):171-176.
Periodontitis in US Adults: National health and nutrition examination
40. Xu X, He J, Xue J, et al. Oral cavity contains distinct niches
survey 2009–2014. J Am Dent Assoc 1939. 2018;149(7):576-588.e6.
with dynamic microbial communities. Environ Microbiol.
20. Kassebaum NJ, Smith AGC, Bernabé E, et al. Global, regional, and
2015;17(3):699-710.
national prevalence, incidence, and disability-adjusted life years
41. Kenney EB, Ash MM. Oxidation Reduction Potential of Developing
for oral conditions for 195 countries, 1990–2015: A systematic
Plaque, Periodontal Pockets and Gingival Sulci. J Periodontol-
analysis for the global burden of diseases, injuries, and risk factors.
Periodontics. 1969;40(11):630-633.
J Dent Res. 2017;96(4):380-387.
42. Diaz PI, Dupuy AK, Abusleme L, et al. Using high throughput se-
21. Frias-Lopez J, Duran-Pinedo A. Effect of periodontal pathogens on
quencing to explore the biodiversity in oral bacterial communities.
the metatranscriptome of a healthy multispecies biofilm model. J
Mol Oral Microbiol. 2012;27(3):182-201.
Bacteriol. 2012;194(8):2082-2095.
43. Kumar PS, Mason MR. Mouthguards: does the indigenous mi-
22. Frias-Lopez J, Shi Y, Tyson GW, et al. Microbial community gene
crobiome play a role in maintaining oral health? Front Cell Infect
expression in ocean surface waters. Proc Natl Acad Sci U S A.
Microbiol. 2015;5:35.
2008;105(10):3805-3810.
|
42       DURAN-PINEDO

44. Mager DL, Ximenez-Fyvie LA, Haffajee AD, Socransky SS. BU063 (Oral Taxon 286) and comparison to the closely related
Distribution of selected bacterial species on intraoral surfaces. J pathogen Tannerella forsythia. PLoS One. 2014;9(2):e89398.
Clin Periodontol. 2003;30(7):644-654. 65. Stewart EJ. Growing unculturable bacteria. J Bacteriol.
45. Proctor DM, Relman DA. The landscape ecology and microbi- 2012;194(16):4151-4160.
ota of the human nose, mouth, and throat. Cell Host Microbe. 66. Kroes I, Lepp PW, Relman DA. Bacterial diversity within
2017;21(4):421-432. the human subgingival crevice. Proc Natl Acad Sci U S A.
46. Ximenez-Fyvie LA, Haffajee AD, Socransky SS. Microbial compo- 1999;96(25):14547-14552.
sition of supra- and subgingival plaque in subjects with adult peri- 67. Sakamoto M, Huang Y, Umeda M, Ishikawa I, Benno Y. Detection
odontitis. J Clin Periodontol. 2000;27(10):722-732. of novel oral phylotypes associated with periodontitis. FEMS
47. Mark Welch JL, Dewhirst FE, Borisy GG. Biogeography of the oral Microbiol Lett. 2002;217(1):65-69.
microbiome: the site-specialist hypothesis. Annu Rev Microbiol. 68. Wade W. Unculturable bacteria—the uncharacterized organisms
2019;73(1):335-358. that cause oral infections. J R Soc Med. 2002;95(2):81-83.
48. Valm AM. The structure of dental plaque microbial communities in 69. de Lillo A, Booth V, Kyriacou L, Weightman AJ, Wade WG.
the transition from health to dental caries and periodontal disease. Culture-independent identification of periodontitis-associated
J Mol Biol. 2019;431(16):2957-2969. Porphyromonas and Tannerella populations by targeted molecular
49. Listgarten MA. Structure of the microbial flora associated with analysis. J Clin Microbiol. 2004;42(12):5523-5527.
periodontal health and disease in man. A light and electron micro- 70. Vartoukian SR, Moazzez RV, Paster BJ, Dewhirst FE, Wade WG.
scopic study. J Periodontol. 1976;47(1):1-18. First cultivation of health-associated Tannerella sp. HOT-286
50. Nyvad B, Fejerskov O. Scanning electron microscopy of early mi- (BU063). J Dent Res. 2016;95(11):1308-1313.
crobial colonization of human enamel and root surfaces in vivo. 71. Beall CJ, Campbell AG, Griffen AL, Podar M, Leys EJ. Genomics
Scand J Dent Res. 1987;95(4):287-296. of the uncultivated, periodontitis-associated bacterium Tannerella
51. Listgarten MA, Mayo HE, Tremblay R. Development of dental sp. BU045 (Oral Taxon 808). mSystems. 2018;3(3):e00018-18.
plaque on epoxy resin crowns in man. A light and electron micro- 72. Ferrari B, Winsley T, Ji M, Neilan B. Insights into the distribution
scopic study. J Periodontol. 1975;46(1):10-26. and abundance of the ubiquitous Candidatus Saccharibacteria
52. Palmer RJ, Shah N, Valm A, et al. Interbacterial adhesion networks phylum following tag pyrosequencing. Sci Rep. 2015;4(1):3957.
within early oral biofilms of single human hosts. Appl Environ 73. Kitten T, Kinscherf TG, McEvoy JL, Willis DK. A newly identi-
Microbiol. 2017;83(11):e00407-17. fied regulator is required for virulence and toxin production in
53. Periasamy S, Chalmers NI, Du-Thumm L, Kolenbrander PE. Pseudomonas syringae. Mol Microbiol. 1998;28(5):917-929.
Fusobacterium nucleatum ATCC 10953 requires Actinomyces 74. Marcy Y, Ouverney C, Bik EM, et al. Dissecting biological “dark
naeslundii ATCC 43146 for growth on saliva in a three-species matter” with single-cell genetic analysis of rare and uncultivated
community that includes Streptococcus oralis 34. Appl Environ TM7 microbes from the human mouth. Proc Natl Acad Sci U S A.
Microbiol. 2009;75(10):3250-3257. 2007;104(29):11889-11894.
54. Egland PG, Palmer RJ, Kolenbrander PE. Interspecies communica- 75. Duran-Pinedo AE, Baker VD, Frias-Lopez J. The periodontal
tion in Streptococcus gordonii-Veillonella atypica biofilms: signal- pathogen Porphyromonas gingivalis induces expression of trans-
ing in flow conditions requires juxtaposition. Proc Natl Acad Sci U S posases and cell death of Streptococcus mitis in a biofilm model.
A. 2004;101(48):16917-16922. Infect Immun. 2014;82(8):3374-3382.
55. Periasamy S, Kolenbrander PE. Aggregatibacter actinomy- 76. Liu B, Faller LL, Klitgord N, et al. Deep sequencing of the oral
cetemcomitans builds mutualistic biofilm communities with microbiome reveals signatures of periodontal disease. PLoS One.
Fusobacterium nucleatum and Veillonella species in saliva. Infect 2012;7(6):e37919.
Immun. 2009;77(9):3542-3551. 77. Brinig MM, Lepp PW, Ouverney CC, Armitage GC, Relman DA.
56. Mark Welch JL, Rossetti BJ, Rieken CW, Dewhirst FE, Borisy GG. Prevalence of bacteria of division TM7 in human subgingival
Biogeography of a human oral microbiome at the micron scale. plaque and their association with disease. Appl Environ Microbiol.
Proc Natl Acad Sci U S A. 2016;113(6):E791-800. 2003;69(3):1687-1694.
57. Wecke J, Kersten T, Madela K, et al. A novel technique for moni- 78. Podar M, Abulencia CB, Walcher M, et al. Targeted access to the
toring the development of bacterial biofilms in human periodontal genomes of low-abundance organisms in complex microbial com-
pockets. FEMS Microbiol Lett. 2000;191(1):95-101. munities. Appl Environ Microbiol. 2007;73(10):3205-3214.
58. Drescher J, Schlafer S, Schaudinn C, et al. Molecular epidemiology 79. He X, McLean JS, Edlund A, et al. Cultivation of a human-associ-
and spatial distribution of Selenomonas spp. in subgingival bio- ated TM7 phylotype reveals a reduced genome and epibiotic para-
films. Eur J Oral Sci. 2010;118(5):466-474. sitic lifestyle. Proc Natl Acad Sci U S A. 2015;112(1):244-249.
59. Marsh PD, Moter A, Devine DA. Dental plaque biofilms: communi- 80. Wang B, Wu J, Lamont RJ, Lin X, Xie H. Negative correlation of dis-
ties, conflict and control. Periodontol 2000. 2011;55(1):16-35. tributions of Streptococcus cristatus and Porphyromonas gingiva-
60. Noiri Y, Ozaki K, Nakae H, Matsuo T, Ebisu S. An immunohisto- lis in subgingival plaque. J Clin Microbiol. 2009;47(12):3902-3906.
chemical study on the localization of Porphyromonas gingivalis, 81. Larsen MH, Leisner JJ, Ingmer H. The chitinolytic activity of
Campylobacter rectus and Actinomyces viscosus in human peri- listeria monocytogenes EGD is regulated by carbohydrates but
odontal pockets. J Periodontal Res. 1997;32(7):598-607. also by the virulence regulator PrfA. Appl Environ Microbiol.
61. Noiri Y, Li L, Ebisu S. The localization of periodontal-disease-as- 2010;76(19):6470-6476.
sociated bacteria in human periodontal pockets. J Dent Res. 82. Jorth P, Turner KH, Gumus P, Nizam N, Buduneli N, Whiteley M.
2001;80(10):1930-1934. Metatranscriptomics of the human oral microbiome during health
62. Noiri Y, Li L, Yoshimura F, Ebisu S. Localization of Porphyromonas and disease. MBio. 2014;5(2):e01012–14.
gingivalis-carrying fimbriae in situ in human periodontal pockets. J 83. Hoeijmakers WAM, Bártfai R, Françoijs K-J, Stunnenberg
Dent Res. 2004;83(12):941-945. HG. Linear amplification for deep sequencing. Nat Protoc.
63. Vartoukian SR. Cultivation strategies for growth of uncultivated 2011;6(7):1026-1036.
bacteria. J Oral Biosci. 2016;58(4):142-149. 84. Duran-Pinedo AE, Yost S, Frias-Lopez J. Small RNA transcriptome
64. Beall CJ, Campbell AG, Dayeh DM, Griffen AL, Podar M, Leys EJ. of the oral microbiome during periodontitis progression. Appl
Single cell genomics of uncultured, health-associated Tannerella Environ Microbiol. 2015;81(19):6688-6699.
DURAN-PINEDO |
      43

85. Hart SN, Therneau TM, Zhang Y, Poland GA, Kocher J-P. predicted by the levels of Porphyromonas gingivalis and
Calculating sample size estimates for RNA sequencing data. J Treponema denticola in subgingival plaque. Oral Microbiol
Comput Biol. 2013;20(12):970-978. Immunol. 2009;24(6):469-477.
86. Ching T, Huang S, Garmire LX. Power analysis and sample size 107. Charalampakis G, Dahlén G, Carlén A, Leonhardt A. Bacterial
estimation for RNA-Seq differential expression. RNA N Y N. markers vs. clinical markers to predict progression of chronic peri-
2014;20(11):1684-1696. odontitis: a 2-yr prospective observational study. Eur J Oral Sci.
87. Liu Y, Zhou J, White KP. RNA-seq differential expression studies: more 2013;121(5):394-402.
sequence or more replication? Bioinformatics. 2014;30(3):301-304. 108. Khalaf H, Lönn J, Bengtsson T. Cytokines and chemokines are
88. Beasley DE, Koltz AM, Lambert JE, Fierer N, Dunn RR. The evo- differentially expressed in patients with periodontitis: possible
lution of stomach acidity and its relevance to the human microbi- role for TGF-β1 as a marker for disease progression. Cytokine.
ome. PLoS One. 2015;10(7):e0134116. 2014;67(1):29-35.
89. He X, McLean JS, Guo L, Lux R, Shi W. The social structure of 109. Heitz-Mayfield LJA. Disease progression: identification of high-
microbial community involved in colonization resistance. ISME J. risk groups and individuals for periodontitis. J Clin Periodontol.
2014;8(3):564-574. 2005;32(Suppl 6):196-209.
90. Kilian M, Chapple ILC, Hannig M, et al. The oral microbi- 110. Ricci M, Garoia F, Tabarroni C, et al. Association between genetic
ome - an update for oral healthcare professionals. Br Dent J. risk score and periodontitis onset and progression: a pilot study.
2016;221(10):657-666. Arch Oral Biol. 2011;56(12):1499-1505.
91. Abubucker S, Segata N, Goll J, et al. Metabolic reconstruction for 111. Socransky SS, Haffajee AD, Goodson JM, Lindhe J. New con-
metagenomic data and its application to the human microbiome. cepts of destructive periodontal disease. J Clin Periodontol.
PLoS Comput Biol. 2012;8(6):e1002358. 1984;11(1):21-32.
92. Benítez-Páez A, Belda-Ferre P, Simón-Soro A, Mira A. Microbiota 112. Haffajee AD, Socransky SS. Attachment level changes in destruc-
diversity and gene expression dynamics in human oral biofilms. tive periodontal diseases. J Clin Periodontol. 1986;13(5):461-475.
BMC Genom. 2014;15:311. 113. Jeffcoat MK, Reddy MS. Progression of probing attachment loss in
93. Kraal L, Abubucker S, Kota K, Fischbach MA, Mitreva M. The prev- adult periodontitis. J Periodontol. 1991;62(3):185-189.
alence of species and strains in the human microbiome: a resource 114. Clerehugh V, Worthington HV, Lennon MA, Chandler R. Site pro-
for experimental efforts. PLoS One. 2014;9(5):e97279. gression of loss of attachment over 5 years in 14- to 19-year-old
94. Marsh PD. In sickness and in health-what does the oral micro- adolescents. J Clin Periodontol. 1995;22(1):15-21.
biome mean to us? An ecological perspective. Adv Dent Res. 115. Sculley DV, Langley-Evans SC. Periodontal disease is associ-
2018;29(1):60-65. ated with lower antioxidant capacity in whole saliva and evi-
95. Raizada MK, Joe B, Bryan NS, et al. Report of the national heart, dence of increased protein oxidation. Clin Sci Lond Engl 1979.
lung, and blood institute working group on the role of microbiota 2003;105(2):167-172.
in blood pressure regulation: current status and future directions. 116. Leggott PJ, Robertson PB, Rothman DL, Murray PA, Jacob RA. The
Hypertension. 2017;70(3):479-485. effect of controlled ascorbic acid depletion and supplementation
96. Lundberg JO, Weitzberg E, Gladwin MT. The nitrate-nitrite-ni- on periodontal health. J Periodontol. 1986;57(8):480-485.
tric oxide pathway in physiology and therapeutics. Nat Rev Drug 117. Singer RE, Buckner BA. Butyrate and propionate: import-
Discov. 2008;7(2):156-167. ant components of toxic dental plaque extracts. Infect Immun.
97. Hyde ER, Andrade F, Vaksman Z, et al. Metagenomic analysis of 1981;32(2):458-463.
nitrate-reducing bacteria in the oral cavity: implications for nitric 118. Szafranski SP, Wos-Oxley ML, Vilchez-Vargas R, et al. High-
oxide homeostasis. PLoS One. 2014;9(3):e88645. resolution taxonomic profiling of the subgingival microbiome for
98. Desvarieux M, Demmer RT, Jacobs DR, et al. Periodontal bacteria biomarker discovery and periodontitis diagnosis. Appl Environ
and hypertension: the oral infections and vascular disease epide- Microbiol. 2015;81(3):1047-1058.
miology study (INVEST). J Hypertens. 2010;28(7):1413-1421. 119. Lamont RJ, Koo H, Hajishengallis G. The oral microbiota: dy-
99. Kurgan S, Kantarci A. Molecular basis for immunohistochemical namic communities and host interactions. Nat Rev Microbiol.
and inflammatory changes during progression of gingivitis to peri- 2018;16(12):745-759.
odontitis. Periodontol 2000. 2018;76(1):51-67. 120. Hajishengallis G, Liang S, Payne MA, et al. Low-abundance biofilm
100. Kistler JO, Booth V, Bradshaw DJ, Wade WG. Bacterial com-
species orchestrates inflammatory periodontal disease through
munity development in experimental gingivitis. PLoS One. the commensal microbiota and complement. Cell Host Microbe.
2013;8(8):e71227. 2011;10(5):497-506.
101. Smith I. Mycobacterium tuberculosis pathogenesis and molecular 121. Hajishengallis G, Darveau RP, Curtis MA. The keystone-pathogen
determinants of virulence. Clin Microbiol Rev. 2003;16(3):463-496. hypothesis. Nat Rev Microbiol. 2012;10(10):717-725.
102. Kostic AD, Gevers D, Pedamallu CS, et al. Genomic analysis iden- 122. Beisel CL, Storz G. Base pairing small RNAs and their roles in global
tifies association of Fusobacterium with colorectal carcinoma. regulatory networks. FEMS Microbiol Rev. 2010;34(5):866-882.
Genome Res. 2012;22(2):292-298. 123. Storz G, Vogel J, Wassarman KM. Regulation by small RNAs in bac-
103. Gur C, Ibrahim Y, Isaacson B, et al. Binding of the Fap2 pro-
teria: expanding frontiers. Mol Cell. 2011;43(6):880-891.
tein of Fusobacterium nucleatum to human inhibitory recep- 124. Michaux C, Verneuil N, Hartke A, Giard J-C. Physiological roles
tor TIGIT protects tumors from immune cell attack. Immunity. of small RNA molecules. Microbiol Read Engl. 2014;160(Pt
2015;42(2):344-355. 6):1007-1019.
104. Goodson JM, Tanner AC, Haffajee AD, Sornberger GC, Socransky 125. Gong H, Vu G-P, Bai Y, et al. A Salmonella small non-coding

SS. Patterns of progression and regression of advanced destruc- RNA facilitates bacterial invasion and intracellular replication
tive periodontal disease. J Clin Periodontol. 1982;9(6):472-481. by modulating the expression of virulence factors. PLoS Pathog.
105. Eley BM, Cox SW. A 2-year longitudinal study of elastase in human 2011;7(9):e1002120.
gingival crevicular fluid and periodontal attachment loss. J Clin 126. Koo JT, Alleyne TM, Schiano CA, Jafari N, Lathem WW. Global
Periodontol. 1996;23(7):681-692. discovery of small RNAs in Yersinia pseudotuberculosis identifies
106. Byrne SJ, Dashper SG, Darby IB, Adams GG, Hoffmann B,
Yersinia-specific small, noncoding RNAs required for virulence.
Reynolds EC. Progression of chronic periodontitis can be Proc Natl Acad Sci. 2011;108(37):E709-E717.
|
44       DURAN-PINEDO

127. Tsai C-H, Liao R, Chou B, Palumbo M, Contreras LM. Genome- 148. Abusleme L, Dupuy AK, Dutzan N, et al. The subgingival microbi-
wide analyses in bacteria show small-RNA enrichment for long and ome in health and periodontitis and its relationship with commu-
conserved intergenic regions. J Bacteriol. 2015;197(1):40-50. nity biomass and inflammation. ISME J. 2013;7(5):1016-1025.
128. Urban JH, Vogel J. Translational control and target recogni-
149. Hajishengallis G. Periodontitis: from microbial immune subversion
tion by Escherichia coli small RNAs in vivo. Nucleic Acids Res. to systemic inflammation. Nat Rev Immunol. 2015;15(1):30-44.
2007;35(3):1018-1037. 150. Chow J, Tang H, Mazmanian SK. Pathobionts of the gastrointes-
129. Caron M-P, Lafontaine DA, Massé E. Small RNA-mediated regula- tinal microbiota and inflammatory disease. Curr Opin Immunol.
tion at the level of transcript stability. RNA Biol. 2010;7(2):140-144. 2011;23(4):473-480.
130. Waters LS, Storz G. Regulatory RNAs in bacteria. Cell.
151. Dabdoub SM, Ganesan SM, Kumar PS. Comparative metagenom-
2009;136(4):615-628. ics reveals taxonomically idiosyncratic yet functionally congruent
131. Sharma CM, Vogel J. Experimental approaches for the discovery communities in periodontitis. Sci Rep. 2016;6:38993.
and characterization of regulatory small RNA. Curr Opin Microbiol. 152. Naik S, Bouladoux N, Wilhelm C, et al. Compartmentalized
2009;12(5):536-546. control of skin immunity by resident commensals. Science.
132. Altuvia S. Identification of bacterial small non-coding RNAs: ex- 2012;337(6098):1115-1119.
perimental approaches. Curr Opin Microbiol. 2007;10(3):257-261. 153. Dalal SR, Chang EB. The microbial basis of inflammatory bowel dis-
133. Sittka A, Sharma CM, Rolle K, Vogel J. Deep sequencing of
eases. J Clin Invest. 2014;124(10):4190-4196.
Salmonella RNA associated with heterologous Hfq proteins in vivo 154. PubMed entry. http://www.ncbi.nlm.nih.gov/pubme​d/11240860.
reveals small RNAs as a major target class and identifies RNA pro- Accessed May 7, 2013.
cessing phenotypes. RNA Biol. 2009;6(3):266-275. 155. Nairz M, Schroll A, Sonnweber T, Weiss G. The struggle for
134. Liu JM, Livny J, Lawrence MS, Kimball MD, Waldor MK, Camilli iron - a metal at the host-pathogen interface. Cell Microbiol.
A. Experimental discovery of sRNAs in Vibrio cholerae by direct 2010;12(12):1691-1702.
cloning, 5S/tRNA depletion and parallel sequencing. Nucleic Acids 156. Fives-Taylor PM, Meyer DH, Mintz KP, Brissette C. Virulence

Res. 2009;37(6):e46. factors of Actinobacillus actinomycetemcomitans. Periodontol.

135. Irnov I, Sharma CM, Vogel J, Winkler WC. Identification 2000;1999(20):136-167.
of regulatory RNAs in Bacillus subtilis. Nucleic Acids Res. 157. Jain S, Darveau RP. Contribution of Porphyromonas gingi-
2010;38(19):6637-6651. valis lipopolysaccharide to periodontitis. Periodontol 2000.
136. Shi Y, Tyson GW, DeLong EF. Metatranscriptomics reveals
2010;54(1):53-70.
unique microbial small RNAs in the ocean’s water column. Nature. 158. Hiroi M, Shimojima T, Kashimata M, et al. Inhibition by

2009;459(7244):266-269. Porphyromonas gingivalis LPS of apoptosis induction in human
137. Murakami S, Fujishima K, Tomita M, Kanai A. Metatranscriptomic peripheral blood polymorphonuclear leukocytes. Anticancer Res.
analysis of microbes in an Oceanfront deep-subsurface hot spring 1998;18(5A):3475-3479.
reveals novel small RNAs and type-specific tRNA degradation. 159. Preshaw PM, Lauffart B, Zak E, Jeffcoat MK, Barton I, Heasman
Appl Environ Microbiol. 2012;78(4):1015-1022. PA. Progression and treatment of chronic adult periodontitis. J
138. Gosalbes MJ, Durbán A, Pignatelli M, et al. Metatranscriptomic ap- Periodontol. 1999;70(10):1209-1220.
proach to analyze the functional human gut microbiota. PLoS One. 160. Murray DA, Wilton JMA. Lipopolysaccharide from the periodon-
2011;6(3):e17447. tal pathogen Porphyromonas gingivalis prevents apoptosis of
139. Romby P, Vandenesch F, Wagner EGH. The role of RNAs in the HL60-derived neutrophils in vitro. Infect Immun. 2003;71(12):
regulation of virulence-gene expression. Curr Opin Microbiol. 7232-7235.
2006;9(2):229-236. 161. Handal T, Olsen I, Walker CB, Caugant DA. Beta-lactamase
140. Soutourina OA, Monot M, Boudry P, et al. Genome-wide identi- production and antimicrobial susceptibility of subgingival bac-
fication of regulatory RNAs in the human pathogen Clostridium teria from refractory periodontitis. Oral Microbiol Immunol.
difficile. PLoS Genet. 2013;9(5):e1003493. 2004;19(5):303-308.
141. Pichon C, Felden B. Small RNA gene identification and 162. van Winkelhoff AJ, Winkel EG, Barendregt D, Dellemijn-Kippuw
mRNA target predictions in bacteria. Bioinforma Oxf Engl. N, Stijne A, van der Velden U. beta-Lactamase producing bacteria
2008;24(24):2807-2813. in adult periodontitis. J Clin Periodontol. 1997;24(8):538-543.
142. Wilderman PJ, Sowa NA, FitzGerald DJ, et al. Identification of 163. Cossart P, Pizarro-Cerdá J, Lecuit M. Invasion of mammalian cells
tandem duplicate regulatory small RNAs in Pseudomonas aeru- by Listeria monocytogenes: functional mimicry to subvert cellular
ginosa involved in iron homeostasis. Proc Natl Acad Sci U S A. functions. Trends Cell Biol. 2003;13(1):23-31.
2004;101(26):9792-9797. 164. Lecuit M, Ohayon H, Braun L, Mengaud J, Cossart P. Internalin
143. Bardill JP, Hammer BK. Non-coding sRNAs regulate virulence in the of Listeria monocytogenes with an intact leucine-rich repeat
bacterial pathogen Vibrio cholerae. RNA Biol. 2012;9(4):392-401. region is sufficient to promote internalization. Infect Immun.
144. Waters JL, Salyers AA. The small RNA RteR inhibits transfer of 1997;65(12):5309-5319.
the bacteroides conjugative transposon CTnDOT. J Bacteriol. 165. Wang C-H, Lin C-Y, Luo Y-H, et al. Effects of oligopeptide
2012;194(19):5228-5236. permease in group a streptococcal infection. Infect Immun.
145. Lee H-R, Rhyu I-C, Kim H-D, et al. In-vivo-induced antigenic deter- 2005;73(5):2881-2890.
minants of Fusobacterium nucleatum subsp. nucleatum. Mol Oral 166. Berezow AB, Darveau RP. Microbial shift and periodontitis.

Microbiol. 2011;26(2):164-172. Periodontol 2000. 2011;55(1):36-47.
146. Mitchell HL, Dashper SG, Catmull DV, et al. Treponema denti- 167. Relman DA. The human microbiome: ecosystem resilience and
cola biofilm-induced expression of a bacteriophage, toxin-an- health. Nutr Rev. 2012;70(Suppl 1):S2-9.
titoxin systems and transposases. Microbiology. 2010;156(Pt 168. Uitto VJ, Suomalainen K, Sorsa T. Salivary collagenase. Origin,
3):774-788. characteristics and relationship to periodontal health. J Periodontal
147. Phillips PL, Progulske-Fox A, Grieshaber NA. Expression of Res. 1990;25(3):135-142.
Porphyromonas gingivalis small RNA in response to hemin avail- 169. Yost S, Duran-Pinedo AE. The contribution of Tannerella forsythia
ability identified using microarray and RNA-seq Analysis. FEMS dipeptidyl aminopeptidase IV in the breakdown of collagen. Mol
Microbiol Lett. 2014;351(2):202-208. Oral Microbiol. 2018;33(6):407-419.
DURAN-PINEDO |
      45

170. Moscoso JA, Schramke H, Zhang Y, et al. Binding of Cyclic Di-AMP 186. Burne RA, Marquis RE. Biofilm acid/base physiology and gene ex-
to the Staphylococcus aureus Sensor Kinase KdpD occurs via the pression in oral bacteria. Methods Enzymol. 2001;337:403-415.
universal stress protein domain and downregulates the expression 187. Edlund A, Yang Y, Yooseph S, et al. Meta-omics uncover temporal
of the Kdp potassium transporter. J Bacteriol. 2016;198(1):98-110. regulation of pathways across oral microbiome genera during in
171. Su J, Gong H, Lai J, Main A, Lu S. The potassium transporter Trk vitro sugar metabolism. ISME J. 2015;9(12):2605-2619.
and external potassium modulate salmonella enterica protein se- 188. Tanner ACR, Kent RL, Holgerson PL, et al. Microbiota of se-

cretion and virulence. Infect Immun. 2009;77(2):667-675. vere early childhood caries before and after therapy. J Dent Res.
172. Tenenbaum H, Jehl F, Gallion C, Dahan M. Amoxicillin and 2011;90(11):1298-1305.
clavulanic acid concentrations in gingival crevicular fluid. J Clin 189. Jakubovics NS. Intermicrobial interactions as a driver for com-
Periodontol. 1997;24(11):804-807. munity composition and stratification of oral biofilms. J Mol Biol.
173. Yost S, Duran-Pinedo AE, Krishnan K, Frias-Lopez J. Potassium is 2015;427(23):3662-3675.
a key signal in host-microbiome dysbiosis in periodontitis. PLOS 190. Kressirer CA, Chen T, Lake Harriman K, et al. Functional pro-
Pathog. 2017;13(6):e1006457. files of coronal and dentin caries in children. J Oral Microbiol.
174. Huynh HTT, Nkamga VD, Drancourt M, Aboudharam G. 2018;10(1):1495976.
Genetic variants of dental plaque Methanobrevibacter ora- 191. Tanner ACR, Mathney JMJ, Kent RL, et al. Cultivable anaero-
lis. Eur J Clin Microbiol Infect Dis Off Publ Eur Soc Clin Microbiol. bic microbiota of severe early childhood caries. J Clin Microbiol.
2015;34(6):1097-1101. 2011;49(4):1464-1474.
175. Huynh HTT, Pignoly M, Drancourt M, Aboudharam G. A new 192. Tanner ACR, Kressirer CA, Rothmiller S, Johansson I, Chalmers NI.
methanogen “Methanobrevibacter massiliense” isolated in a case The caries microbiome: implications for reversing dysbiosis. Adv
of severe periodontitis. BMC Res Notes. 2017;10(1):657. Dent Res. 2018;29(1):78-85.
176. Guo L, Hu W, He X, Lux R, McLean J, Shi W. investigating acid pro- 193. Peterson SN, Meissner T, Su AI, et al. Functional expression of
duction by Streptococcus mutans with a surface-displayed pH-sen- dental plaque microbiota. Front Cell Infect Microbiol. 2014;4:108.
sitive green fluorescent protein. PLoS One. 2013;8(2):e57182. 194. May A, Brandt BW, El-Kebir M, et al. metaModules identifies
177. Zeng L, Burne RA. Sucrose- and fructose-specific effects on the key functional subnetworks in microbiome-related disease.
transcriptome of Streptococcus mutans, as determined by RNA Bioinformatics. 2016;32(11):1678-1685. https://doi.org/10.1093/
sequencing. Appl Environ Microbiol. 2016;82(1):146-156. bioinformatics/btv526. Epub 2015 Sep 5.PMID: 26342232
178. Marsh PD. Microbial ecology of dental plaque and its significance 195. Kilian M, Poulsen K, Blomqvist T, et al. Evolution of Streptococcus
in health and disease. Adv Dent Res. 1994;8(2):263-271. pneumoniae and its close commensal relatives. PLoS One.
179. Simón-Soro A, Tomás I, Cabrera-Rubio R, Catalan MD, Nyvad 2008;3(7):e2683.
B, Mira A. Microbial geography of the oral cavity. J Dent Res. 196. Loo CY, Mitrakul K, Voss IB, Hughes CV, Ganeshkumar N.
2013;92(7):616-621. Involvement of an inducible fructose phosphotransferase op-
180. Simón-Soro A, Belda-Ferre P, Cabrera-Rubio R, Alcaraz LD, Mira eron in Streptococcus gordonii biofilm formation. J Bacteriol.
A. A tissue-dependent hypothesis of dental caries. Caries Res. 2003;185(21):6241-6254.
2013;47(6):591-600. 197. Muto M, Hitomi Y, Ohtsu A, Ebihara S, Yoshida S, Esumi H.
181. Gross EL, Leys EJ, Gasparovich SR, et al. Bacterial 16S sequence Association of aldehyde dehydrogenase 2 gene polymorphism
analysis of severe caries in young permanent teeth. J Clin Microbiol. with multiple oesophageal dysplasia in head and neck cancer pa-
2010;48(11):4121-4128. tients. Gut. 2000;47(2):256-261.
182. Johansson I, Witkowska E, Kaveh B, Lif Holgerson P, Tanner ACR.
The microbiome in populations with a low and high prevalence of
caries. J Dent Res. 2016;95(1):80-86.
How to cite this article: Duran-Pinedo AE. Metatranscriptomic
183. Simón-Soro A, Mira A. Solving the etiology of dental caries. Trends
Microbiol. 2015;23(2):76-82.
analyses of the oral microbiome. Periodontol 2000.
184. Nyvad B, Machiulskiene V, Baelum V. Reliability of a new caries di- 2021;85:28–45. https://doi.org/10.1111/prd.12350
agnostic system differentiating between active and inactive caries
lesions. Caries Res. 1999;33(4):252-260.
185. Peterson SN, Snesrud E, Liu J, et al. The dental plaque microbiome
in health and disease. PLoS One. 2013;8(3):e58487.

You might also like