Open access peer-reviewed chapter

Understanding Oral Diseases: Exploring Opportunities from Filipino Oral Microbiome Research

Written By

Marilen P. Balolong and Michael Antonio F. Mendoza

Submitted: 26 May 2020 Reviewed: 26 October 2020 Published: 08 February 2021

DOI: 10.5772/intechopen.94751

From the Edited Volume

Dental Caries

Edited by Efka Zabokova Bilbilova

Chapter metrics overview

687 Chapter Downloads

View Full Metrics

Abstract

The human mouth houses the second most diverse microbial community in the body, with almost 700 species of bacteria colonizing the hard surfaces of teeth and the soft tissues of the oral mucosa. To compete in the relatively exposed oral cavity, resident microbes must avoid being replaced by newcomers. This selective constraint, coupled with pressure on the host to cultivate a beneficial microbiome, has rendered a commensal oral microbiota that displays colonization resistance, protecting the human host from invasive species, including pathogens. Current control of dental plaque-related diseases is non-specific and is centered on the removal of plaque by mechanical means. Several new methods based on the modulation of the microbiome that aim at maintaining and re-establishing a healthy oral ecosystem have been developed and has greatly expanded our knowledge of the composition and function of the oral microbiome in health and disease. Promoting a balanced microbiome is therefore important to effectively maintain or restore oral health. This review provides an updated body of knowledge on oral microbiome in health and disease and discusses the implications for modern-day oral healthcare. Filipino Oral Microbiome Research to develop a policy framework for microbiome-based management of dental diseases and opportunities will be discussed.

Keywords

  • oral microbiome
  • dysbiosis
  • modulation
  • systemic diseases
  • oral health policy

1. Summary

Resident microorganisms have co-evolved and co-existed in our body in a mostly harmonious symbiotic relationship. The mouth, for instance, houses the second most diverse microbial community in the body, with almost 700 species of bacteria colonizing the hard surfaces of teeth and the soft tissues of the oral mucosa. Synergy and interaction of variable oral microorganisms help human body against invasion of undesirable stimulation outside. To compete in the relatively exposed oral cavity, resident microbes must avoid being replaced by newcomers. This selective constraint, coupled with pressure on the host to cultivate a beneficial microbiome, has rendered a commensal oral microbiota that displays colonization resistance, protecting the human host from invasive species, including pathogens. Perturbations of the oral microbiome through modern-day lifestyles can have detrimental consequences for our general and oral health. During dysbiosis, the equilibrium of the oral ecosystem is disrupted, allowing disease-promoting bacteria to manifest and cause conditions such as caries, gingivitis and periodontitis. In addition, rapid increases in carbohydrate consumption have disrupted the evolved homeostasis between the oral microbiota and dental health, as reflected by the high prevalence of dental caries. Development of novel modalities to prevent caries has been the subject of a breadth of research.

The current control of dental plaque-related diseases is nonspecific and is centered on the removal of plaque by mechanical means. The most prevalent oral diseases, dental caries and periodontal diseases, are microbiota-associated diseases. Due to this realization about the oral microbiome, several new methods based on the modulation of the microbiome that aim at maintaining and reestablishing a healthy oral ecosystem have been developed. Oral microbiomes play an important role in the human microbial community and human health. The use of recently developed molecular methods has greatly expanded our knowledge of the composition and function of the oral microbiome in health and disease. Studies in oral microbiomes and their interactions with microbiomes in variable body sites and variable health condition are critical in our cognition of our body and how to make effect on human health improvement. Through recent advances in technology, complexities of the oral microbiome have been slowly unraveled and new insights into its role during both health and disease are now made available. For practitioners and patients alike, promoting a balanced microbiome is therefore important to effectively maintain or restore oral health.

This chapter aims to give an update on our current knowledge of the oral microbiome in health and disease and to discuss implications for modern-day oral healthcare. More importantly, opportunities and future directions tapping Filipino Oral Microbiome Research will be explored toward developing a research-driven policy framework for microbiome-based management of dental diseases.

Advertisement

2. The oral microbiome: an overview

Humans have co-evolved with microorganisms [1], these commensals prevent colonization by exogenous microorganisms which translates their significant involvement in the normal development of the host defenses and gut mucosa, during the production of vitamin and energy as well as in the regulation of the cardiovascular system [2]. It was Antony van Leeuwenhoek who was first to identify oral microorganisms using his microscope, which is possibly the first account of oral microbiome [3], his “animalcules” [4].

The term “oral microbiome” has been coined to describe the microbial communities inhabiting the oral cavity behaving as a consortium involved in the modulation of various pathophysiological conditions of the host [5, 6, 7]. It is considered as the second most complex of the human body, after the gut-associated microbiome [8, 9, 10] with the average adult harboring about 50 to 100 billion bacteria represent about 200 predominant bacterial species [11]. Compared to other body sites, the oral microbiome is unique and readily accessible. However, there are only about 700 predominant taxa: 54% are cultivable and identified; 14% are cultivable but not yet identified, and the remaining 32% not even cultivated yet, the so-called “uncultivables” or phylotypes [5, 11, 12, 13]. These estimations are based on years of traditional identification of bacteria from cultural and phenotypic characterization studies, but mostly from identification of bacteria from culture-independent molecular studies using 16S rRNA gene comparative analyses [5, 14, 15, 16, 17].

Figure 1 describes the transition and changes related microbiome profile in the mouth from sterility to oral disease state such as dental caries or systemic disease state. The fetus in the womb is usually sterile [18, 19]. The baby comes in contact with the microflora of the uterus and vagina of the mother during delivery, and later with the microorganisms of the atmosphere at birth [20]. The oral cavity of the newborn is considered sterile even though there is a high probability of contracting contaminants. Since the mouth is exposed to various microorganisms from the first feeding onward, this starts the process of acquiring the resident oral microflora [19]. Typically, the earliest colonizers of the tooth surface are commensal streptococci, such as Streptococcus mitis, Streptococcus sanguinis, Streptococcus gordonii, and other closely related taxa [21]. They antagonize transient microorganisms by producing by-products such as alkali, bacteriocins, and hydrogen peroxides. High abundances of these commensal streptococci are favored in dental plaque particularly in the absence of a carbohydrate-rich diet. This dominance is strongly associated with good dental health.

Figure 1.

Transition and changes related microbiome profile in the mouth from sterility to oral disease state such as dental caries or systemic disease state.

Historically, humans recorded two shifts in diet preference as influenced by the development of agriculture and by the industrial revolution [22] which favored the increase of carbohydrate consumption, particularly sucrose. With frequent consumption of carbohydrates such as sucrose coupled with poor oral hygiene, bacterial production of a certain glucan matrix is enhanced which can lead to the development of dental plaque, where other carbohydrate-related fermentation can continue. Due to the described changes that would result, perturbation of homeostasis of the oral microbiome can now lead to the development of dental caries, which is considered as the most common chronic disease worldwide, affecting 60–90% of children and adults even in industrialized countries [23].

It was previously mentioned that about 400 to 500 oral taxa have been detected in the subgingival crevice alone [14, 16] with the remaining taxa distributed to other oral habitats such as the tongue, tooth surface, buccal mucosa, tonsils, soft and hard palate, and lip vestibule [14, 24, 25, 26]. Although there is compositional variation between sample sites taken from the oral cavity, a ‘core’ microbiome in health has been identified [24, 25, 27]. Studies have also demonstrated that oral disease is not due to an isolated organism such as Streptococcus mutans causing caries, but is more polymicrobial in nature [5, 28, 29]. Studies have identified Bifidobacterium, Veillonella, Granulicatetta, Scardovia, Fusobacterium, Prevotella and Actinomyces as potential contributors to early childhood caries evidenced by their altered abundance in the caries microbiota [30].

Interestingly, there is a wide range of microorganisms that are said to inhabit the human oral cavity, including bacteria, fungi, viruses, archaea and protozoa which likewise contribute to influencing oral and systemic health [31]. Bacteria account for the main portion of oral microorganisms, and the major knowledge of the composition of oral bacteria comes from past culture-dependent methods, however, these data substantially underestimated the composition of the oral microbiome but development of culture-independent methods, particularly targeting 16S ribosomal RNA, offered expanded awareness of the great richness and diversity of the oral microbiome [31]. Fungi are present widely in the oral cavity not only as opportunistic pathogens of the elderly and immune-compromised but may also be present as members of the healthy oral microbiota [32]. Archaea constitutes only a minor part of the oral microbiome and is restricted to limited species [33, 34] observed in healthy subjects, but their prevalence and numbers are elevated in individuals with periodontitis [6]. Most viruses in the mouth are related to diseases including herpes simplex virus, human papilloma virus [35] and HIV infection indirectly related to observed oral manifestations, such as oral candidiasis, oral hairy leukoplakia, linear gingival erythema and necrotizing ulcerative periodontitis, and Kaposi’s sarcoma [36].

The salivary or oral microbiome has been the target of interest for its diagnostic and prognostic value [37]. Since the commensal microbiome has an important role in the maintenance of oral and systemic health, altering its delicate balance may lead to the development of certain oral pathologies such as cavities endodontic disease, periodontal diseases, osteitis and tonsillitis [38, 39, 40] as well as various systemic diseases including cardiovascular disease [41], obesity [42, 43], heart disease [44], diabetes [45], pediatric Crohn’s Disease [46], pancreatic cancer [47], colon carcinoma [48], and even psychiatric disorders [49]. Studies during the last decade focused on the management and prevention of dental caries by modulating oral microbiome [37, 50]. However, it is yet to be established if there is a causal relationship between the oral microbiome and these systemic disorders. This battleground represents a significant opportunity for intervention and subsequent prevention of caries.

Advertisement

3. Methods employed in studying the oral microbiome: from “culturomics” to whole genome sequencing

Tables 1 and 2 show a comprehensive list of methods used in the study of oral microbiome from culture-dependent method to whole genome sequencing and OMICS. The relevant findings from the studies that utilized the various methods are also presented. Historically, as guided by Koch’s postulates, cultivable bacterial taxa associated with various oral diseases were identified using culture-dependent methodologies such as microscopy, biochemical and other phenotypic tests, growth conditions, sugar utilization, and antibiotic sensitivity. However, this approach has presented various limitations when it comes to describing the actual diversity of the oral microbiome, or the so-called “great plate anomaly” [94, 95] and is unable to fully characterize complexity of bacterial communities such as those found in the oral cavity [60, 96].

Methods to study oral microbiomeStudies that have utilized the given methodsImportant findings
Culture-Dependent Approach: “Culturomics”
Heart Infusion Blood Agar (with menadione)
Mitis Salivarius Medium (MS)
[51]The cultivation on rich medium under anaerobic conditions, they investigated the overall composition of the microbiota of saliva, dental plaque, the gingival crevice, the cheeks, and the tongue dorsum
They found organisms such as Corynebacterium spp., Actinomyces spp., and Streptococcus sanguinis colonizing teeth, treponemes in the gingival crevice, and Streptococcus salivarius on the tongue
MM10 Sucrose Medium
MSB (MS + 20% sucrose, 0.2 U/ml bacitracin) Medium
[52]Significant association between plaque levels of S. mutans and caries
Saliva samples tended to have low levels of S. mutans and were equivocal in demonstrating a relationship between I and caries
MM10 Sucrose Medium
MSB Medium
LBS Medium
[53]Clinical decay can occur in a few instances in the absence of detectable S. mutans, as was observed in the fissures high in lactobacilli
Trypticase Soy Broth (TSB) with 1% glucose[54]Streptococcus mutans, Lactobacillus casei and Streprococcus faecalis showed greater acid tolerance than strains of Streptococcus sanguis, Streptococcus salivarius, Streptococcus mitis and Actinomyces viscosus
Species of plaque bacteria most closely associated with the initiation or progression of dental caries are more aciduric than non-cariogenic species
MSB medium
GSTB medium (5% glucose, 5% sucrose, telurite, and bacitracin)
TYCSB medium (tryptone, yeast extract, cysteine, sucrose, and bacitracin)
[55]Early culture-based studies had shown that enamel caries was associated with increases in the numbers and proportions of mutans streptococci
Blood agar
BM Medium
[56]If such conditions of low pH are repeated on a regular basis, then the acidogenic/aciduric species are eventually able to increase their proportions and drive the plaque pH even lower, outcompeting the beneficial species
Tidd Hewitt Broth[57]There is heterogeneity in terms of expression of attributes among clinical strains belonging to a species, so that some strains of mutans streptocococci can be less acidogenic than isolates of other streptococcal species
Emphasizes the need for detailed species and biovar identification of oral streptococci and for recognition of the significant physiological differences that occur within single species
BM Medium
BMHGM Medium (BM supplemented with hog gastric mucin)
[58]If such conditions of low pH are repeated on a regular basis, then the acidogenic/aciduric species are eventually able to increase their proportions and drive the plaque pH even lower, outcompeting the beneficial species
MTPY Medium
TYCSB Medium
[59]A total of 424 bifidobacteria were identified and these were Bifidobacterium dentium, Parascardovia denticolens, Scardovia inopicata, Bifidobacterium longum, Scardovia genomosp. C1 and Bifidobacterium breve
Suggested that bifidobacteria may play a role in the progression of occlusal caries lesions in both children and adults
Recent culture-based studies have correlated more diverse communities of bacteria with caries, including reporting on the association of Actinomyces and Bifidobacterium species with lesions, often with mutans streptococci comprising a relatively small percentage of the microbiota at diseased sites
BHI + HK Medium
FAA Medium
BUA medium
[5]The human oral cavity contains a number of different habitats, including the teeth, gingival sulcus, tongue, cheeks, hard and soft palates, and tonsils, which are colonized by bacteria
More than 700 prokaryotic taxa have been detected in the oral cavity, many of which cannot be isolated by common culture methods
Actinobacteria, Bacteroidetes, Chlamydiae, Chloroflexi, Euryarchaeota, Firmicutes, Fusobacteria, Proteobacteria, Spirochaetes, SR1, Synergistetes, Tenericutes, and TM7
Cooked Meat Medium (CMM)
CMM with mucin and serum
Blood Agar
[60]Cultivation of a Synergistetes strain representing a previously uncultivated strain from Subgingival plaque
Blood Agar
MacConkey’s Medium
Chapman’s Plate Growth Medium
Chromagar-Candida
[61]Samples deriving from oral cavity swabs, cultures, and in vitro tests showed the presence of various microorganisms belonging to different genera, species, and strains of bacteria, protozoans, and fungi in patient groups analyzed
The pretreatment examination of oral cavity microbiota may be helpful in a preventive approach to the spread of infectious microorganisms, which may be etiological agents of human opportunistic infections and risk factors for treatment complications, particularly dangerous for older adults

Table 1.

Comprehensive list of methods used in the study of oral microbiome from culture-dependent methods to whole genome sequencing and OMICS. The relevant findings from the studies that utilized the methods are also presented.

Culture-Independent Approach: Pre-Next Generation Sequencing Era to Whole Genome Sequencing
Studies that have utilized culture-independent methodsMethods usedImportant findings
[14]ABI Prism cycle sequencing kitHighly common genera in various regions of cavity:
Gemella, Granulicatella, Streptococcus, and Veillonella
[62]16S ribosomal RNA genes polymerase chain reaction (PCR)Tumor specimens: Exiguobacterium oxido- tolerans, Prevotella melaninogenica, Staphylococcus aureus, Veillonella parvula
Non-tumor specimens: Moraxella osloensis, Prevotella veroralis, Actinomyces
*Oral squamous cell carcinoma
[63]Comparative 16S rRNA gene sequencing and QPCRDental Plaque: Microbial community is dominated by Streptococci (66%) followed by Actinomyces (6%)
[64]16S ribosomal RNA genes polymerase chain reaction (PCR)
Restriction fragment length polymorphism
16S ribosomal RNA gene sequencing
L. salivarius was more prevalent in children with moderate to high caries prevalence compared with children with low caries prevalence, while L. fermentum was the most predominant species in all study groups.
Genetic heterogeneity of Lactobacillus species was found among the children and those with high caries prevalence tended to be colonized with more than one clonal type
L. salivarius may be a putative caries pathogen among preschool Thai children
[27]454 Pyrosequencing FLX systemDental surfaces, cheek hard palate, tongue and saliva:
Firmicutes: Streptococcus, Veillonellaceae, Granulicatella
Proteobacteria: Neisseria, Haemophilus
Actinobacteria: Corynebacterium, Rothia, Actinomyces
Bacteroidetes: Prevotella, Capnocytophaga, Porphyromonas
[65]Sanger SequencingGingiva: 247 species-level phylotypes and nine bacterial phyla
[5]ABI Prism cycle sequencing kitVarious Regions of the Oral Cavity:
A total of 619 taxa in 13 phyla Actinobacteria, Bacteroidetes,
Chlamydiae, Chloroflexi, Euryarchaeota, Firmicutes, Fusobacteria
Proteobacteria, Spirochaetes, SR1, Synergistetes, Tenericutes, and TM7
[60]Fluorescent in situ Hybridization (FISH)
Quantitative PCR (Q-PCR)
Synergistetes W090/OTU 6.2 to be prevalent and ‘abundant’ in both periodontally healthy subjects and those with chronic periodontitis, which would suggest that this bacterium is a commensal, endogenous to the oral cavity
[66]454 Pyrosequencing FLX systemSupragingival and subgingival plaques: Five major phyla are Bacteroidetes, Actinobacteria, Firmicutes, Proteobacteria and Fusobacteria
[67]Arbitrarily primed PCR (AP-PCR)
Chromosomal DNA fingerprinting
DGGE
Ascertained the role of lactobacilli in the caries process
Found seven LB species in the oral cavity of the subjects: L. vaginalis, L. oris, L. gasseri, L. salivarius, L. fermentum, L. rhamnosus and L. casei.
[68]16S rRNA-based microarray and PCRSeveral species, including S. wiggsiae and S. exigua, are associated with the ecology of advanced caries
Successful treatment is accompanied by a change in the microbiota, and that severe early childhood caries is diverse, with influences from selected bacteria or from diet
[69]454 GLX Titanium pyrosequencingResults show a given bacterial consortium associated with cariogenic and non-cariogenic conditions, in agreement with the existence of a healthy oral microbiome and giving support to the idea of dental caries being a polymicrobial disease
[70]454 Pyrosequencing using GS- FLX sequencerSupragingival dental plaque:
Healthy one: Bacilli and Gam- maProteobacteria dominated with specific association of Rothia and Aggregatibacter
Diseased one: Clostridiales and Bacteroidetes dominated
[71]Next generation sequencing of 16S rRNA gene
DNA deduction
Data included 9 phyla, 16 classes, 26 orders, 55 families, and 111 genera (OUT was defined within 3% genetic difference)
Detected approximately 29% more types of microbes than those detected from the same sample without using DNA deduction
DNA deduction technique will lead to a better understanding of the diversity of the human oral microbiota
[72]DGGE and Sanger sequencingOral Tissue:
Six major phyla: Firmicutes, Bacteroidetes Proteobacteria, Fusobacteria, Actinobacteria and uncultivated TM7 with 80 bacterial species/phylotypes
[73]PCR-DGGEFirst study that provides a baseline profile of the oral microbial diversity in caries-free and caries-active Filipino adults using culture- independent techniques
The caries-free group exhibited a more diverse microflora compared with its caries-active
[74]Next generation sequencing 16S rRNA whole genome (454 FLX Titanium)Saliva microbiomes in human population were featured by a vast phylogenetic diversity yet a minimal organismal core
Caries microbiomes were significantly more variable in community structure whereas the healthy ones were relatively conserved
Abundance changes of certain taxa such as overabundance of Prevotella Genus distinguished caries microbiota from healthy ones
Caries-active and normal individuals carried different arrays of Prevotella species
No ‘caries-specific’ operational taxonomic units (OTUs) were detected, yet 147 OTUs were ‘caries associated’, that is, differentially distributed yet present in both healthy and caries-active populations
[75]RNA-Seq Paired-end sequencing (Illumina HiSeq)The bacteria changing activity during biofilm formation and after meal ingestion were person-specific
Some individuals showed extreme homeostasis with virtually no changes in the active bacterial population after food ingestion, suggesting the presence of a microbial community which could be associated to dental health
[76]16S pyrotag sequencingSubgingival Plaque:
Smokers: elevated number of S. mutans, Lactobacillus sali varius and commensal poor anaerobes
[77]Roche 454 FLX sySwab:
Significant reduction in Firmicutes and Actinobacteria while Fusobacteria count proportionally elevated in all oral cancer patients
[78]Arbitrary-primed PCR
(AP-PCR)
Multi-locus Sequence Analysis (MLSA)
Develop a streamlined method for identifying strains of S. oralis and S. mitis from plaque samples so that they could be analyzed in a separate study devoted to low pH streptococci and caries
Novel primer sets offer a convenient means of presumptive identification that will have utility in many studies where large scale, in-depth genomic analyses are not practical
[79]RNA-Seq Paired-end sequencing (Illumina HiSeq)There were similar levels of Actinomyces gene expression in both sound and carious root biofilms
These bacteria can be commensal in root surface sites but may be cariogenic due to survival mechanisms that allow them to exist in acid environments and to metabolize sugars, saving energy
[80]PhyloChip microarraysOral buccal mucosa:
IBS-overweight participants showed decreased richness in the phylum Bacteroidetes
[81]454 Pyrosequencing FLX systemOral rinse samples:
13 phyla having 122 genera Core microbiome constituted 7 phyla with 55 genera
[82]454-pyrosequencingSubgingival plaque:
Diabetics: Fusobacterium, Parvimonas, Peptostreptococcus
Gemella, Streptococcus, Leptotrichia, Filifactor, Veillonella, TM7, Terrahemophilus and elevated levels of Capnocytophaga
Pseudomonas, Bergeyella, Sphingomonas, Corynebacterium
Propionibacterium, and Neisseria in hyperglycemic individuals
[83]Deep shotgun sequencingSaliva from three pairs of populations of hunter-gatherers and traditional farmers living in close proximity in the Philippines
Comparing these microbiomes with publicly available data from individuals living on a Western diet revealed that abundance ratios of core species were significantly correlated with subsistence strategy, with hunter-gatherers and Westerners occupying either end of a gradient of Neisseria against Haemophilus, and traditional farmers falling in between
Species found preferentially in hunter-gatherers included microbes often considered as oral pathogens, despite their hosts’ apparent good oral health
[84]Next-generation sequencing of V3-V4 region of 16S rRNA gene (Illumina MiSeq)Salivary microbiome was characterized in a group of children stratified by the Simplified Oral Hygiene Index (OHI-S)
Twenty taxonomic groups (Seventeen genera, two families and one class; Streptococcus, Veillonella, Gemellaceae, Prevotella, Rothia, Porphyromonas, Granulicatella, Actinomyces, TM-7-3, Leptotrichia, Haemophilus, Selenomonas, Neisseria, Megasphaera, Capnocytophaga, Oribacterium, Abiotrophia, Lachnospiraceae, Peptostreptococcus, and Atopobium) were found in all subjects and constituted 94.5–96.5% of the microbiome
Of these twenty genera, the proportion of Streptococcus decreased while Veillonella increased with poor oral hygiene status, Veillonella dispar and Veillonella parvula tended to be elevated in the Poor oral hygiene group
[85]Next-generation sequencing of V4 region of 16S rRNA gene (Illumina MiSeq)A comprehensive analysis of the oral microbiome identified Granulicatella and Neisseria as bacteria enriched in subjects with MetS and Peptococcus as bacteria abundant in healthy controls
Results support that local oral microbiota can be associated with systemic disorders
The microbial biomarkers identified would aid in determination of which individuals develop chronic diseases from their MetS and contribute to strategic disease management
[86]Next-generation sequencing of V1-V2 region of 16S rRNA gene (Illumina MiSeq)Porphyromonas, Treponema, Tannerella, Filifactor, and Aggregatibacter were more abundant in patients with periodontal disease, whereas Streptococcus, Haemophilus, Capnocytophaga, Gemella, Campylobacter, and Granulicatella were found at higher levels in healthy controls
[87]Fluorescence in situ hybridization and Confocal Laser Scanning MicroscopyEstablishes S. oralis as commensal keeper of homeostasis in the biofilm by antagonizing S. mutans, thus preventing a caries-favoring dysbiotic state
[88]Next-generation sequencing of V3-V4 region of 16S rRNA gene (Illumina MiSeq)Interproximal-associated microbiota was found to be similar to already described bacterial communities in other mouth niches. Streptoccocus, Veillonella, Rothia, Actinomyces, Neisseria, Haemophilus and Fusobacterium were the most abundant genera in this oral region
[30]Next-generation sequencing of V4-V5 region of 16S rRNA gene (Illumina MiSeq)Distinct differences between the caries microbiota and saliva microbiota were identified, with separation of both salivary groups (caries-active and caries-free)
The major phyla of the caries active dentinal microbiota were Firmicutes (median abundance value 33.5%) and Bacteroidetes (23.2%), with Neisseria (10.3%) being the most abundant genus, followed by Prevotella (10%).
The caries-active salivary microbiota was dominated by Proteobacteria (median abundance value 38.2%) and Bacteroidetes (27.8%) with the most abundant genus being Neisseria (16.3%), followed by Porphyromonas (9.5%).
Caries microbiota samples were characterized by high relative abundance of Streptococcus mutans, Prevotella spp., Bifidobacterium and Scardovia spp.
[89]Next-generation sequencing of V3-V4 region of 16S rRNA gene (Illumina MiSeq)Provides thorough knowledge of the microbiological etiology of elderly individuals with caries and is expected to provide novel methods for its prevention and treatment
[90]Next-generation sequencing of V3-V4 region of 16S rRNA gene (Illumina MiSeq)Decrease in commensal saliva bacteria were observed in all the body sizes when compared to normal weight children. Notably, the relative abundance of bacteria related to, Veillonella, Prevotella, Selenomonas, and Streptococcus was reduced in obese children
Body size–specific saliva microbiota profiles open new avenues for studying the potential roles of microbiota in weight development and management
[91]Fluorescence in situ hybridization and Confocal Laser Scanning MicroscopyInvestigated the impact of various Fusobacterium species on in vitro biofilm formation and structure in three different oral biofilm models namely a supragingival, a supragingival “feeding”, and a subgingival biofilm model
Study showed variations in the growing capacities of different fusobacteria within biofilms, affecting the growth of surrounding species and potentially the biofilm architecture
[92]Next-generation sequencing of V4 region of 16S rRNA gene (Illumina MiSeq)Adult oral microbiomes were predominantly impacted by oral health habits, while youth microbiomes were impacted by biological sex and weight status
The oral pathogen Treponema was detected more commonly in adults without recent dentist visits and in obese youth
Oral microbiomes from participants of the same family were more similar to each other than to oral microbiomes from non-related individuals
[8]Whole Genome Sequencing
Real-time quantitative PCR microarray
Sampled oral micro-habitat included tongue dorsum, hard palate, buccal mucosa, keratinized gingiva, supragingival and subgingival plaque, and saliva with or without rinsing. Each sampled oral niche evidenced a different microbial community, including bacteria, fungi, and viruses
Oral rinse microbiome was more representative of the whole site-specific microbiomes, compared with that of saliva
Healthy oral microbiome resistome included highly prevalent genes conferring resistance to macrolide, lincosamides, streptogramin, and tetracycline.
[93]Next-generation sequencing of V3-V4 region of 16S rRNA gene (Illumina MiSeq)A comparison of oral bacteriome between two groups revealed the dominance of acidogenic and aciduric bacteria in diabetics which suggested the involvement of these eubacteria in oral dysbacteriosis in diabetes mellitus
Phylum Firmicutes (p-value = 0.024 at 95% confidence interval) was significantly more abundant among diabetic patients than among the controls
Acidogenic bacteria Prevotella (p-value = 0.024) and Leptotrichia (p-value = 1.5 x 10−3); and aciduric bacteria Veillonella (p-value = 0.013) were found to be in higher abundance in diabetic patients

Table 2.

Comprehensive list to describe methods in the study of oral microbiome from culture-dependent methods to whole genome sequencing and OMICS + studies that have utilized hem through the years).

Conventional cultivation of oral bacteria samples requires taking an appropriate sample, transferring the sample to an appropriate medium for transportation, and followed by correct storage following collection. Dispersion and plating the bacteria onto various culture media in the laboratory will then follow. The bacteria are then isolated and characterized by their colony morphologies (appearance) and biochemical testing. Species which do not grow are naturally “overlooked”. One of the major challenges facing oral microbiology is the ability to culture the yet-to-be cultivated 32% of oral species. Furthermore, current knowledge existing about pathogens linked to oral diseases are based on bacterial communities that have somehow survived transportation in a sample, grew easily or rapidly in the laboratory, based on a culture method employed in the laboratory [96, 97] which seems to limited and fundamentally incomplete.

Resultant knowledge of oral microbial ecology and dynamics have enabled development of novel culture techniques that have somehow helped the cultivation of some strains including the provision of distinct conditions such as an anaerobic (oxygen-free) environment, incubation in a variety of temperatures, use of chemically-defined media containing specific amounts of nutrients, and even the use of cytokine networks and microbial co-colonizers [98, 99]. But despite these advances, still many organisms remained uncultivable due to several assumptions [98, 100] such as (1) they exist in obligate metabolic associations with other organisms; (2) since oral bacteria do not live in isolation but in complex communities called biofilms, they depend on synergies and antagonisms for growth, along with mutual reliance for growth and survival; and (3) the lack of essential nutrients, growth factors and/or signaling molecules, overfeeding conditions, lack of cross-feeding partners, culture media toxicity and disruption of bacterial quorum-sensing and other signaling systems or other reasons that remained to be undiscovered until now.

Cultivation remains the foundation of oral microbiology in characterizing phenotypically, identification, including physiology and pathogenicity of particular species. The proportion therefore of the so-called ‘overlooked’ bacteria that are responsible for a number of oral diseases [99] must be studied using molecular signatures in order to include a huge number of uncultivable species found in healthy and diseased sites in the human mouth and to better understand the many aspects of microbial dynamics in the oral cavity [95, 98, 101]. The development and advances in molecular techniques, therefore, can help enhance and enable the study of complex host-associated bacterial communities such as those in the oral cavity.

Watson and Crick’s discovery of the DNA structure helped clarify the mechanism of base pairing and explained how genetic information is stored and copied in living organisms. This knowledge then led to the power to investigate microbial communities or individual cells from detection to identification to diversity profiling using molecular-based technologies. Molecular techniques have enabled a more in-depth investigation and have resulted in different and more focused approaches compared to cultivation [102].

Early high-throughput analytical approaches in studying microbial communities utilized the fragment size separation differences of the DNA such as the denaturing gradient gel electrophoresis (DGGE) and the restriction fragment length polymorphism (RFLP) methods which based findings on [103, 104] following amplification of target regions of interest through the polymerase chain reaction or PCR. These types of approach enabled analysis at the macro-level for microbial communities characterized by large shifts or variations in the population. DNA–DNA hybridization and DNA microarrays followed providing rapid assessment of specific bacterial associations in oral health and disease [105] with the use of hybridized DNA fragments and complementary probes arrayed on a glass slide for expression profiling [106].

The modern era of microbial genome analysis began in the early 2000s, with metagenomics and gene sequencing techniques [107]. The 16S rRNA gene is usually selected as the target gene because it comprises both variable and conserved regions, permitting the use of primers to conserved regions and more specific primers to amplify 16S rRNA genes from any source to discriminate between taxa [108, 109]. Bacterial taxa can be phylogenetically identified, whether they are cultivable or not-yet-cultivated, in a mixed population, e.g., plaque, by isolating DNA, amplification of universally conserved primers for 16S rRNA gene, a conserved gene of approximately 1500 bp, followed by cloning into Escherichia coli, and lastly Sanger sequencing [16] or the more modern next generation sequencing [110]. Among nine distinct hypervariable regions, V1, V2, V3 and V4 stretches are highly exploited sequences for studying microbial diversity due to their extreme variability; V5 exhibits least variability [111] while V4–6 region is considered as the most reliable stretch that represents the entire length of 16S rDNA for studying the majority of bacterial phyla [110]. Currently, oral microbiome-based next generation sequencing analysis chiefly relies on primers of either V1–2 or V3–4 regions. These bits and pieces of 16S rDNA regions are capable enough of providing the entire picture of bacterial phyla present in a niche; however, ambiguous data obtained from various targeted variable regions cannot be ignored, and it demands a massive full- length sequencing of 16S rDNA region for its validation. Interestingly, the use of V1 region has been linked to differentiating Streptococci, a pioneer of the oral cavity, while region V2 is associated to accurately identify various phylotypes of Gram-negative Porphyromonas and Fusobacterium [111]. Furthermore, amplifying V1–3 works best forStreptococcus, Fusobacterium, Prevotella, Porphyromonas, and Bacteroides, but the use of V4–6 region showed observed dominance of Prevotella, Porphyromonas, Treponema, Enterococci and Campylobacter-like oral inhabitants [112]. In the last decade, next generation sequencing methods have revolutionized the study of microbial diversity, and enabled large-scale sequencing projects to be completed in a few days or sometimes hours.

The use of universal bacterial primers for simultaneous analysis of a wide variety of bacteria in the oral microbiota is undeniably extensive. Some limitations related to its possible binding to the same conserved area of the 16S rRNA target population have been reported and cannot be ignored such as those linked to probable PCR competitive inhibition. In this scenario, the DNA of the predominant (major) bacterial species is much more likely to be amplified than DNA from bacteria that form a minority or small proportion of the overall mixed population which could lead (in theory) to the complete omission of the DNA of minority bacterial species from the analysis [113]. Amplification of the DNA species present in excess (competitor DNA) may result at the expense of the DNA species present in smaller amounts limiting the analysis to only the majority species present. To address this, Kuwamura and Kamiya [114] developed the “DNA deduction” approach where excess DNA of the majority bacterial species present in the target population is removed and the detection of minority bacterial members of the community is facilitated. This method was proved to be a useful technique to better understand the diversity and composition of the human oral microbiota since many researchers identify species based on 16S rRNA sequence information in studying the oral microbiota [115, 116]. Since a large number of good-quality sequences is required in order to construct a precise DNA database for use in human microbiota analyses for next generation sequencing, DNA deduction technique may offer improvement of the human microbiota database.

Molecular techniques based on next-generation sequencing (NGS) of the 16 rRNA gene of the bacterial genome, allowed analysis of the complexity of the bacterial component of the oral microbiome and has represented the standard for studying the composition of microbial communities by allowing differentiation of bacteria by sequencing the variable regions of the gene coding for the 16S ribosomal RNA (rRNA) which greatly improved our knowledge of the bacterial component of the oral microbiome [13, 117]. However, it does not usually provide sufficient information to resolve communities at the sub-species level, nor it can detect eukaryotic microorganisms and/or viruses. Instead, the species-level resolution obtained by NGS is not adequate for transmission studies or for exploring subspecies variation in disease association, and the oral microbiome includes also important non-bacterial components, including eukaryotic microbes (fungi, protozoa) and viruses [8]. For instance, reports on normal microbiome have been almost exclusively restricted to the bacteriome, and there are limited published findings on the mycobiome–fungal microbiome and on other microorganisms. To simultaneously characterize the presence and amount of all the microbial components potentially present in the oral cavity, the Whole Genome Sequencing (WGS) was introduced very recently [118, 119]. In whole genome sequencing, the entire DNA (genome) of a single microbial culture or a complex microbial population can now be sequenced to great depth allowing us to generate reference genomes (de novo assembly) as a resource for future studies or identify the composition of microbial community respectively (mapping back to a reference genome).

Another important aspect that may not also be addressed in depth is the issue related to antimicrobial resistance or AMR, very limited reports are available on resistome of the healthy oral microbiome [120, 121]. Since AMR is a growing concern, it would be useful to have data on the prevalence and type of drug resistance of the microbes composing the healthy oral microbiome, which might be very easily acquired and transmitted through aerosol and contact. Recently, Caselli et al. [8] provided comprehensive and detailed picture of the healthy oral microbiome as determined by WGS analysis, including also the drug-resistance features of the bacterial component. These findings further strengthened laboratory capabilities and added more areas for oral microbiome research to enable evidence-based oral diseases management in practice.

Advertisement

4. Impacts of oral microbiome to oral health (dental caries) and non-oral health (systemic diseases)

Oral microbiome data have accumulated in recent years and somehow slowly influences how we see management of oral diseases. During the last decade, studies have focused on the management and prevention of certain and disorders such as dental caries by modulating oral microbiome [37, 50]. Interestingly, some member of the oral microbiota has also been tagged as possible effective biosensors or biomarkers of oral or systemic diseases [84, 122]. In addition, the salivary or oral microbiome has been the target of interest for its diagnostic and prognostic value [37]. Table 3 comprehensively lists the studies that supported the impacts of oral microbiome studies to both Oral and Non-Oral or Systemic Health.

Oral microbiota derived from microbiome dataAssociated disease statusReferences
Fusobacterium nucleatumPeriodontal disease[123]
Eubacterium minutum
Prevotella intermedia
Peri-implantitis[124]
HaemophilusOral leukoplakia (mucosal disease)[125]
Fusobacteria
Leptotrichia spp
Campylobacter concisus
[126]
Prevotella spp.
Lactobacillus spp.
Dialister spp.
Filifactor spp.
Pathogenesis and progression of dental caries[127]
Veillonella
Porphyromonas
Streptococcus mutans
Severe early childhood caries[128]
Streptococcus
Porphyromonas
Actinomyces
Ma et al., 2015
Streptococcus
Prevotella
Neisseria
Haemophilus
Veillonella
Gemella
Inflammatory Bowel Disease[129]
Klebsiella spp.Atarashi et al., 2017
Acinetobacter calcoaceticus
Atopobium rimae
Clavibacter michiganensis subsp. tessellarius
Bacillus mycoides
Capnocytophaga gingivalis
Citrobacter koseri
Curtobacterium flaccumfaciens
Delftia acidovorans
Eikenella corrodens isolate
Escherichia coli
Enterococcus faecalis
Lactobacillus gasseri
Fusobacterium canifelinum, Fusobacterium naviforme, Fusobacterium nucleatum ssp. 1 nucleatum
Gemella haemolysans, Gemella morbillorum
Leptotrichia shahii
Megasphaera micronuciformis
Moraxella osloensis
Ralstonia insidiosa, Ralstonia pickettii, Ralstonia solanacearum
Rothia
Novel Atopobium
Olsenella uli
Plantibacter flavus
Propionibacterium acnes
Parvimonas
Peptostreptococcus micros, Peptostreptococcus stomatis
Porphyromonas
Prevotella melaninogenica
Rhodococcus erythropolis
Rothia mucilaginosa
Slackia
Streptococcus gordoniii, Streptococcus salivarius, Streptococcus sanguinis
Tepidimonas aquatica
Thermus scotoductus
Oral Squamous Cell Carcinoma[130]
[131]
[132]
[72]
[133]
[134]
[7]
Porphyromonas gingivalis
Aggregatibacter actinomycetemcomitans
Leptotrichia
Pancreatic Cancer[135]
[136]
[39]
StreptococcusPancreatic Ductal Adenocarcinoma[137]
Rothia
Peptostreptococcus
Fusobacterium
Leptotrichia
Pancreatic Head Cancer[138]
Porphyromonas gingivalisGingival Squamous Cell Carcinoma[139]
Rothia mucilaginosa
Lactobacillus gasseri
Lactobacillus johnsonii
Lactobacillus gavinalis
Streptococcus salivarius
Streptococcus vestibularis
Fusobacterium nucleatum
Head and Neck Squamous Cell Carcinoma[140]
Rothia mucilaginosa
Capnocytophaga gingivalis
Prevotella melininogenica
Gemella morbillorum
Granulicatella adiacens
Streptococcus gordonii
Streptoccus parasanguinis
Stretococcus salivarius
Fusobacterium nucleatum
Oral Potentially Malignant Disorder[141]
Gemella morbillorumKeratocytic Odontogenic Tumor[142]
Streptococcus
Fusobacterium
Colorectal Carcinoma[143]
Porphyromonas gingivalis
Prevotella
Streptococcus
Esophageal Squamous Cell Carcinoma[144, 145]
StreptococcusGastric Adenocarcinoma[146]
Prevotella melaninogenica
Fusobacterium
Oral Cancer[77]
Rothia mucilaginosa
Streptococus
Oral Mobile Tongue Carcinoma[147]
Actinobacillus actinomycetemcomitans
Tannerella forsythia
Porphyromonas gingivalis
Fusobacterium nucleatum
Prevotella intermedia
Alzheimer’s disease[148, 149]
Aggregatibacter
Neisseria
Gemella
Eikenella
Selenomonas
Actinomyces
Capnocytophaga
Fusobacterium
Veillonella
Streptococcus
Diabetes[150]
Bacteroides forsythus
Campylobacter rectus
Adverse pregnancy outcomes[151]
Fusobacterium nucleatum[152]
Lactobacillus salivariusRheumatoid arthritis[153]
Veillonella
Prevotella
Megasphaera
Campylobacter
Human Immunodeficiency Virus infection[154]
Chryseomonas
Veillonella
Streptococcus
Atherosclerosis[155]

Table 3.

Comprehensive list of studies supporting the impacts of Oral microbiome data (healthy or dysbiosis) to Oral and non-Oral health.

Dental diseases are now viewed as a consequence of a deleterious shift in the balance of the normally stable resident oral microbiome. It is known that frequent carbohydrate consumption or reduced saliva flow can lead to caries, and excessive plaque accumulation increases the risk of periodontal diseases. In general, increase in fermentable carbohydrates in the oral cavity or saliva, which is usually the case in diabetes, establishes a favorable environment for the microbes involved in dental caries [13]. However, reports have also described that shifts in the proportion of plaque microflora may not be directly caused by the mere availability of certain fermentable carbohydrates but rather, brought about by pH-mediated mediated reactions generated from carbohydrate metabolism [56]. Hence, it can be suggested that modifying metabolism of certain carbohydrates may result in slowing down the enrichment of cariogenic species, preventing development of caries. The excess uptake of carbohydrates leads to acid production due to fermentation of carbohydrates by many oral cavity inhabitants which disturbs the buffering capacity of saliva and hence results in tooth decay or dental caries.

Changes in environmental conditions brought about by poor oral hygiene may favor certain members of the oral microbiota and consequently serve as biomarkers for certain oral disease condition. Saliva and diet were observed to alter Lactobacillus abundance in the pulpal layer [156], and that low levels of Lactobacillus were linked to the degree and duration of pain and length of caries destruction. Moreover, poor oral hygiene status seems to be linked to anaerobic conditions in the salivary milieu, this condition inhibits growth of Streptococcus and favors the growth of Veillonella [153, 157, 158]. Salivary and plaque microbiome data also revealed that Veillonella was frequently found in subjects with caries or periodontal subjects, whether children or adults, with poor oral health and having caries or periodontitis [84]. Based on these findings, the proportion of Veillonella in salivary microbiome, therefore, can serve as a biological indicator for poor oral hygiene and caries in children and adults.

Oral microbial dysbiosis is also linked to oral inflammation and may contribute to systemic conditions through bacteremia [159]. It contributes to variable systemic diseases processing including gastrointestinal system diseases like inflammatory bowel disease, liver cirrhosis, pancreatic cancer, nervous system diseases like Alzheimer’s disease, endocrine system diseases like diabetes, adverse pregnancy outcomes, obesity and polycystic ovary syndrome, immune system diseases like rheumatoid arthritis and HIV infection, and cardiovascular system diseases like atherosclerosis [160].

The dominant genera, Streptococcus, Prevotella, Neisseria, Haemophilus, Veillonella and Gemella, were found to largely contribute to dysbiosis observed in the salivary microbiota of IBD patients [129]. A growing interest regarding the variation of salivary microbiota between dental caries patients with comorbidities such as rheumatoid arthritis (RA) and atherosclerosis compared to healthy subjects. Alterations in the gut, dental or saliva microbiome distinguished individuals with RA from healthy controls, were correlated with clinical measures and could be used to stratify individuals on the basis of their response to therapy. It was observed that Haemophilus species seems to be depleted from saliva, dental plaque and fecal samples of RA patients but their numbers become normal upon standard RA treatment [153]. Interestingly, the levels of Porphyromonas gingivalis, Treponema denticola, Tannerella forsythia and Prevotella intermedia in the saliva, have been linked in lowering serum levels of high-density lipoproteins, which may be associated with an increased risk of atherosclerosis [122]. These findings also highlight the possibility of saliva-based screening as alternative to fecal samples in microbiologic studies of systemic diseases.

Microbes are believed to be released from the biofilm through the epithelium and spread systemically via the blood circulation, this is probably the reason why bacteria isolated from pancreatic tissues are believed to be possible members of the oral microbiome [161]. Porphyromonas gingivalis is an important bacterium that can be transferred from the mouth to gut in many diseases, including colon cancer, IBD, and diabetes [138]. This microorganism induces dysbiosis by impairing innate host defenses while promoting inflammatory responses in phagocytic cells. Since this microbe can disrupt the interaction between host microbiota and mucosa by modulating the innate immune system and signaling pathway enhancing high levels of inflammation in pancreatic cancer [162], it is believed to be a good candidate biosensor for early diagnosis of pancreatic cancer. Likewise, oral metabolome finds its application in determination of biomarkers in oral cancers [163]. An elevated rate of degradation of macromolecules was shown in metabolome profiling in periodontal disease. Shifts in metabolic profiles indicate survival of pathogens in given conditions.

Disruption of the oral microbiome leads to dysbiosis. Identifying the microbiome in health is the first step of human microbiome research, after which it is necessary to understand the role of the microbiome in the alteration of functional and metabolic pathways associated with diseased states [9]. Salivary microbiota composition and abundance were significantly associated with body size and dependent on gender; particularly notable was the decrease in the core bacteria in overweight and obese children [90]. Overweight and obese children are likely to stay obese into adulthood and develop diseases more frequently than normal weight children. Thus, the early identification of subjects at risk of developing obesity and the prevention of overweight and obesity is of great importance. As we increase our understanding of the interplay between the environment and the oral microbiome, it will become possible to identify new strategies to combat disease by actively promoting our natural microbiota and reducing the impact of the drivers of dysbiosis [164].

Future studies focusing on precision medicine and risk prediction for dental caries and even periodontitis may also be possible when changes in oral microbiota profiles in the salivary milieu may be made available. Previous studies have supported this which targeted subtypes or strains of specific bacterial species such as Porphyromonas gingivalis or Aggregatibacter actinomycemcomitans [165] in periodontits or Streptococcus mutans in caries [166] in predicting common oral health risks. Characterizing oral biofilms by metabolic activity rather than by listing the predominant species may also be considered as a logical approach when defining plaque biofilms in health and disease.

Advertisement

5. Novel microbiome-based approaches to prevention and treatment of dental caries

Traditional intervention methods to addressing dental diseases include mechanical debridement and antibiotic use. Mechanical means are non-specific and may remove beneficial bacteria in the process, more importantly, the microbial richness and biodiversity are also significantly decreased after mechanical debridement [167] which is not beneficial to overall oral health. Antibiotics on the other hand are designed to target specific pathogenic bacteria in animals and humans [168]. It was reported that the number of amoxicillin-resistant oral bacteria was significantly higher in young children with amoxicillin use than that of children without [169]. In view of the role of oral microorganisms in the causation and pathogenesis of oral and systemic diseases, it is crucial to improve oral protection against pathogens and maintain the dynamic equilibrium of the oral microecology. Understanding the interactions between the microbial communities is a key to combating oral pathogens. Novel strategies have been developed, such as the use of probiotics and prebiotics to address limitations of traditional intervention methods.

Probiotics are well-known in health promotion, they are “live microorganisms when administered in adequate amounts, confer a health benefit on the host” [170]. The mechanism of action of probiotics in the mouth is presumed to be similar to those observed in other parts of the body [171] which is to act mainly through these paths: competition with potential pathogens for nutrients or adhesion sites, killing or inhibition of growth of pathogens through production of bacteriocins or other products, improvement of intestinal barrier integrity and upregulation of mucin production, modulation of cell proliferation and apoptosis, and stimulation and modulation of the mucosal immune system [171, 172]. Oral probiotics should have specific characteristics to perform effectively including the ability to stick to and colonize oral tissue including hard, non-shedding surfaces and become a part of the biofilm [171, 173]. Additionally, they should not ferment sugars; otherwise, they will decrease pH and develop caries [171]. Probiotic methods have been studied to treat caries mainly by interfering with the oral colonization of cariogenic pathogens. Some strains that were used include Lactobacillus casei [174], L. rhamnosus GG [175], L. rhamnosus, Bifidobacterium [176], Lactobacillus reuteri [177], B. animalis [178], and L. paracasei [179], all of which have been verified to be able to decrease the number of cariogenic bacteria and thus, prevent dental caries.

Prebiotics, on the otherhand are poorly digested oligosaccharides and have been demonstrated to be an aid to complement probiotics in the treatment of oral diseases [172] by stimulating the growth and activity of beneficial bacteria and simultaneously inhibiting the growth and activity of potentially detrimental bacteria [172]. Some examples are Lactose, Inulin, Fructo oligosacccharides, Galactooligosaccharides and Xylooligosaccharides [180]. However, studies on prebiotic utilization seems limited and should be further investigated. Based on these findings, probiotics and prebiotics could be alternatives to prevent and cure bacterial diseases because they can reestablish an ecological balance or regain the biodiversity of oral microbiota in its early stages [181]. Prebiotics can drive beneficial changes in the oral microbiota and could increase resistance to dysbiosis and recovery of health. However, interaction between the oral microbiome and probiotics, as well as the exact mode of action of oral probiotics should be taken into serious consideration.

Other promising alternatives to control dental caries are the use of avirulent S. mutans produced by genetic engineering [182], which was designed to target glucosyltransferases and consequently inhibit biofilm formation [183].

The use of bacteriophage or phage is a virus that specifically targets and destroys disease-causing bacteria by invading bacterial cells [182], disrupting their metabolism and causing lysis such as phages against Enterococcus faecalis that showed reduction in bacterial viability in infected root canals [184]. Several targeted delivery systems have been designed and developed to treat oral diseases including nanoparticles [185, 186, 187].

Modulation of oral microbiota, therefore, in combination with novel drugs and delivery approaches serves as promising interventions and opportunities [188]. The application of ecological principles can help us understand how the tight interplay of the oral microbiota and the host dictates health or disease. We encourage advancing research directed toward developing ways or strategies to shift from traditional treatment to preventive and personalized dentistry.

Advertisement

6. Future directions for Philippine-based research and policy: oral microbiome and dental diseases in focus

The oral cavity is inhabited by hundreds of bacterial species that play vital roles in maintaining oral health or in shifting to a diseased state such as dental caries. The observed bacterial profile of the Filipinos compared with other populations may be brought about by the difference in their diet or oral hygiene practices. However, the possibility of this correlation has not been covered in a previous study on the Filipino oral microbiome [73]. Other factors influencing the shifts in the oral microbial diversity from a healthy to a diseased state may also be the focus of future studies.

The continued advancement of research and the resulting robust data on the oral microbiome is expected to lead to evidence-informed dentistry. The link of oral health diseases to systemic conditions have been established. Effective preventive measures, accurate diagnosis, sound treatment and the maintenance of good oral health will be expected outcomes when updated information on the oral microbiome are utilized optimally and may also lead to a decrease of the global burden of non-communicable diseases such as heart disease and diabetes mellitus. This strengthens the call for enhanced research on the Filipino oral microbiome and to shift the perspective that knowledge of the oral microbiome transcends health of the oral cavity but the general well-being of the individual and the populace.

Addressing the 87.4% dental caries and 48.3% periodontal disease prevalence [189] among Filipinos will not only require a responsive oral health care system but behavioral changes for Filipinos to prioritize oral health. The Lifecycle Approach has been laid out by the Department of Health in the delivery of the Basic Package of Oral Health Care [190]. Preventive Services must be done to provide specific protection from the occurrence of dental caries and other dental diseases, and this is initiated by the careful checking of the oral cavity.

Kits to test salivary pH levels are currently available. With its results analyzed with dietary patterns and oral hygiene habits, an individual’s caries risk can be assessed. Minimum clinical intervention are done when appropriate preventive measures are adopted. Diagnostic tests to assess the oral microbiome were not specified in the DOH guidelines but utilization of these test kits, when available, can aid in the development of individualized preventive programs. It should be noted that though the dental public health implications of diagnostic tests may be limited at the moment, its genomic value may be of promise. The high prevalence of oral diseases in the Philippines necessitates that unmet clinical needs be provided with proper preventive and promotive interventions. The development of a Filipino Oral Microbiome Database may support the advancement of effective oral health treatment options which could support responsive oral health policies in establishing oral health programs for Filipinos.

Figure 2 describes our proposed framework directing possible future directions in Philippine-based Research and Policy Focusing on Oral Microbiome and Dental Caries. It should be emphasized that monitoring and evaluation must be included in all stages. The process must be dynamic enough to allow for review. All stakeholders must be open to conducting new research when the desired outcomes are not being realized. With the enactment of the Universal Health Care (UHC) Act in 2018, the State is mandated to provide a comprehensive and integrated health care model to all Filipinos that include cost-effective promotive and preventive services. The translational data from oral microbiome researches can be referred for Health Technology Assessment to be reviewed for inclusion in the oral health care packages under the UHC.

Figure 2.

Proposed framework directing possible future directions in Philippine-based research and policy focusing on Oral microbiome and dental caries.

References

  1. 1. Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biology. 2016;14(8):e1002533
  2. 2. Relman DA. The human microbiome: Ecosystem resilience and health. Nutrition Reviews. 2012;70(Suppl 1):S2-S9
  3. 3. Yamashita Y, Takeshita T. The oral microbiome and human health. Journal of Oral Science. 2017;59:201-206
  4. 4. Patil S, Rao RS, Amrutha N, Sanketh DS. Oral microbial flora in health. World J Dent. 2013;4:262-266
  5. 5. Dewhirst FE, Chen T, Izard J, Paster BJ, Tanner AC, Yu WH, et al. The human oral microbiome. Journal of Bacteriology. 2010;192:5002-5017
  6. 6. He J, Li Y, Cao Y, Xue J, Zhou X. The oral microbiome diversity and its relation to human diseases. Folia Microbiologica. 2015;60(1):69-80
  7. 7. Zhao H, Chu M, Huang Z, Yang X, Ran S, Hu B, et al. Variations in oral microbiota associated with oral cancer. Sci Rep-Uk. 2017;7(1):11773
  8. 8. Caselli E, Fabbri C, D’Accolti M, Soffritti BC, Mazzacane S, Franchi M. Defining the oral microbiome by whole-genome sequencing and resistome analysis: The complexity of the healthy picure. BMC Microbiology. 2020;20:120
  9. 9. Deo PN, Deshmukh R. Oral microbiome: Unveiling the fundamentals. J Oral Maxillofac Pathol. 2019;23(1):122-128
  10. 10. Verma D, Garg PK, Dubey AK. Insights into the human oral microbiome. Archives of Microbiology. 2018;200:525-540
  11. 11. Krishnan K, Chen T, Paster BJ. A practical guide to the oral microbiome and its relation to health and disease. Oral Diseases. 2017;23(3):276-286
  12. 12. Backhed F, Fraser CM, Ringel Y, Sanders ME, Sartor RB, Sherman PM, et al. Defining a healthy human gut microbiome: Current concepts, future directions, and clinical applications. Cell Host & Microbe. 2012;12(5):611-622
  13. 13. Wade WG. The oral microbiome in health and disease. Pharmacological Research. 2013;69(1):137-143
  14. 14. Aas JA, Paster BJ, Stokes LN, Olsen I, Dewhirst FE. Defining the normal bacterial flora of the oral cavity. Journal of Clinical Microbiology. 2005;43:5721-5732
  15. 15. Aas JA, Griffen AL, Dardis SR, Lee AM, Olsen I, Dewhirst FE, et al. Bacteria of dental caries in primary and permanent teeth in children and young adults. Journal of Clinical Microbiology. 2008;46:1407-1417
  16. 16. Paster BJ, Boches SK, Galvin JL, Ericson RE, Lau CN, Levanos VA, et al. Bacterial diversity in human subgingival plaque. Journal of Bacteriology. 2001;183:3770-3783
  17. 17. Paster BJ, Olsen I, Aas JA, Dewhirst FE. The breadth of bacterial diversity in the human periodontal pocket and other oral sites. Periodontology 2000. 2006;42:80-87
  18. 18. Batabyal B, Chakraborty S, Biswas S. Role of oral microflora in human population: A brief review. Int J Pharm Life Sci. 2012;3:2220-2227
  19. 19. Marsh PD. Role of the oral microflora in health. Microbiol Ecol Health Dis. 2009;12:130-137
  20. 20. Sampiao-Maia B, Monteiro-Silva F. Acquisition and maturation of oral microbiome throughout childhood: An update. Dent Res J (Isfahan). 2014;11:291-301
  21. 21. Baker JL, Edlund A. Exploiting the Oral microbiome to prevent tooth decay: Has evolution already provided the best tools? Frontiers in Microbiology. 2019;9:3323
  22. 22. Adler CJ, Dobney K, Weyrich LS, Kaidonis J, Walker AW, Haak W, et al. Sequencing ancient calcified dental plaque shows changes in oral microbiota with dietary shifts of the Neolithic and industrial revolutions. Nature Genetics. 2013;45:450-455
  23. 23. Pitts NB, Zero DT, Marsh PD, Ekstrand K, Weintraub JA, Ramos-Gomez F, et al. Dental caries. Nature Reviews. Disease Primers. 2017;3:17030
  24. 24. Human Microbiome Project C. A framework for human microbiome research. Nature. 2012; 486:215–21. [PubMed: 22699610
  25. 25. Human Microbiome Project C. Structure, function and diversity of the healthy human microbiome. Nature. 2012c; 486:207–14. [PubMed: 22699609]
  26. 26. Segata N, Haake SK, Mannon P, Lemon KP, Waldron L, Gevers D, et al. Composition of the adult digestive tract bacterial microbiome based on seven mouth surfaces, tonsils, throat and stool samples. Genome Biology. 2012;13:R42
  27. 27. Zaura E, Keijser BJF, Huse SM, Crielaard W. Defining the healthy “core microbiome” of oral microbial communities. BMC Microbiology. 2009;9:259
  28. 28. Gross EL, Leys EJ, Gasparovich SR, Firestone ND, Schwartzbaum JA, Janies DA, et al. Bacterial 16S sequence analysis of severe caries in young permanent teeth. Journal of Clinical Microbiology. 2010;48(11):4121-4128
  29. 29. Nyvad B, Crielaard W, Mira A, Takahashi N, Beighton D. Dental caries from a molecular microbiological perspective. Caries Research. 2013;47:89-102
  30. 30. Hurley E, Mullins D, Barrett MP, O’Shea CA, Kinirons M, Anthony Ryan C, et al. The microbiota of the mother at birth and its influence on the emerging infant oral microbiota from birth to 1 year of age: A cohort study. Journal of Oral Microbiology. 2019;11:1
  31. 31. Zhang Y, Wang X, Li H, Ni C, Du Z, and Yan F. 2018. Human oral microbiota and its modulation for oral health. Biomed Pharmacother (Biomedecine pharmacotherapie) 99:883–893.
  32. 32. Krom BP, Kidwai S, Ten Cate JM. Candida and other fungal species: Forgotten players of healthy oral microbiota. Journal of Dental Research. 2014;93(5):445-451
  33. 33. Dridi B, Raoult D, Drancourt M. Archaea as emerging organisms in complex human microbiomes. Anaerobe. 2011;17(2):56-63
  34. 34. Nguyen-Hieu T, Khelaifia S, Aboudharam G, Drancourt M. Methanogenic archaea in subgingival sites: A review. APMIS. 2013;121(6):467-477
  35. 35. Kumaraswamy KL, Vidhya M. Human papilloma virus and oral infections: An update. Journal of Cancer Research and Therapeutics. 2011;7(2):120-127
  36. 36. Reznik DA. Oral manifestations of HIV disease. Top HIV Med. 2006;13:143-148
  37. 37. Lazarevic V, Whiteson K, Gaïa N, Gizard Y, Hernandez D, Farinelli L, et al. Analysis of the salivary microbiome using culture-independent techniques. Journal of Clinical Bioinformatics. 2012;2(1):4
  38. 38. Abusleme L, Dupuy AK, Dutzan N, Silva N, Burleson JA, Strausbaugh LD, et al. The subgingival microbiome in health and periodontitis and its relationship with community biomass and inflammation. The ISME Journal. 2013;7(5):1016-1025
  39. 39. Fan X, Alekseyenko AV, Wu J, Peters BA, Jacobs EJ, Gapstur SM, et al. Human oral microbiome and prospective risk for pancreatic cancer: A population-based nested case-control study. Gut. 2018;67:120-127
  40. 40. Kerr AR. The oral microbiome and cancer. Journal of Dental Hygiene. 2015;89(Suppl 1):20-23
  41. 41. Beck JD, Offenbacher S. Systemic effects of periodontitis: Epidemiology of periodontal disease and cardiovascular disease. Journal of Periodontology. 2005;76(11 Suppl):2089-2100
  42. 42. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: Human gut microbes associated with obesity. Nature. 2006;444(7122):1022-1023
  43. 43. Turnbaugh, P. J. et al. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Science Translational Medicine 1, 6ra14–6ra14 (2009).
  44. 44. Leishman SJ, Do HL, Ford PJ. Cardiovascular disease and the role of oral bacteria. Journal of Oral Microbiology. 2010. DOI: 10.3402/jom.v2i0.5781
  45. 45. Demmer RT, Jacobs DR Jr, Singh R, Zuk A, Rosenbaum M, Papapanou PN, and Desvarieux M. 2015. Periodontal bacteria and prediabetes prevalence in origins: the oral infections, glucose intolerance, and insulin resistance study. J Dent Res.94:201S – 211S.
  46. 46. Docktor MJ, Paster BJ, Abramowicz S, Ingram J, Wang YE, Correll M, et al. Alterations in diversity of the oral microbiome in pediatric inflammatory bowel disease. Inflammatory Bowel Diseases. 2012;18:935-942
  47. 47. Farrell JJ, Zhang L, Zhou H, Chia D, Elashoff D, Akin D, et al. Variations of oral microbiota are associated with pancreatic diseases including pancreatic cancer. Gut. 2012;61:582-588
  48. 48. Ahn J, Sinha R, Pei Z, Dominianni C, Wu J, Shi J, et al. Human gut microbiome and risk for colorectal cancer. Journal of the National Cancer Institute. 2013;105(24):1907-1911
  49. 49. Foster JA, McVey Neufeld KA. Gut-brain axis: How the microbiome influences anxiety and depression. Trends in Neurosciences. 2013;36(5):305-312
  50. 50. Acharya A, Chan Y, Kheur S, Jin LJ, Watt RM, Mattheos N. Salivary microbiome in non-oral disease: A summary of evidence and commentary. Archives of Oral Biology. 2017;83:169-173
  51. 51. Gibbons RJ. Bacteriology of dental caries. Journal of Dental Research. 1964;43(6):1021-1028
  52. 52. Loesche WJ, Rowan J, Straffon LH, Loos PJ. Association of Streptococcus mutans with dental decay. Infection and Immunity. 1975;11(6):1252-1260
  53. 53. Loesche WJ, Straffon LH. Longitudinal investigation of the role of Streptococcus mutans in human fissure decay. Infection and Immunity. 1979;26(2):498-507
  54. 54. Harper DS, Loesche WJ. Growth and tolerance of human dental plaque bacteria. Archives of Oral Biology. 1984;29(10):843-848
  55. 55. Loesche WJ. Role of Streptococcus mutans in human dental decay. Microbiological Reviews. 1986;50(4):353-380
  56. 56. Bradshaw DJ, McKee AS, Marsh PD. The use of defined inocula stored in liquid nitrogen for mixed-culture chemostat studies. J Microbiol Meth. 1989;9:123-128
  57. 57. Kneist S, Scharff S, de Soet JJ, van Loveren C, Stosser L. Bacteriocin production by human strains of mutans and oral streptococci. Caries Res. 2000;34:308
  58. 58. Bradshaw DJ, Homer KA, Marsh PD, Beighton D. Metabolic cooperation in oral microbial communities during growth on mucin. Microbiology. 1994;140(Pt 12):3407-3412
  59. 59. Mantzourani M, Gilbert SC, Sulong HNH, Sheehy EC, Tank S, Fenlon M, et al. The isolation of bifidobacteria from occlusal carious lesions in children and adults. Caries Research. 2009;43:308-313
  60. 60. Vartoukian SR, Palmer RM, Wade WG. Cultivation of a Synergistetes strain representing a previously uncultivated lineage. Environmental Microbiology. 2010;12:916-928
  61. 61. Zawadzki PJ, Perkowski K, Padzik M, Mierzwińska-Nastalska E, Szaflik JP, Conn DB, et al. Examination of oral microbiota diversity in adults and older adults as an approach to prevent spread of risk factors for human infections. BioMed Research International. 2017;2017:8106491
  62. 62. Hooper SJ, Crean SJ, Lewis MAO, Spratt DA, Wade WG, Wilson MJ. Viable bacteria present within oral squamous cell carcinoma tissue. Journal of Clinical Microbiology. 2006;44(5):1719-1725
  63. 63. Dalwai F, Spratt DA, Pratten J. Use of quantitative PCR and culture methods to characterize ecological flux in bacterial biofilms. Journal of Clinical Microbiology. 2007;45(9):3073-3076
  64. 64. Piwat S, Teanpaisan R, Thitasomakul S, Thearmontree A, Dahlén G. Lactobacillus species and genotypes associated wih dental caries in Thai preschool children. Molecular Oral Microbiology. 2010;25(2):157-164
  65. 65. Bik EM, Long CD, Armitage GC, Loomer P, Emerson J, Mongodin EF, et al. Bacterial diversity in the oral cavity of 10 healthy individuals. The ISME Journal. 2010;4:962-974
  66. 66. Xie G, Chain PSG, Lo CC, Liu KL, Gans J, Merritt J, et al. Community and gene composition of a human dental plaque microbiota obtained by metagenomic sequencing. Molecular Oral Microbiology. 2010;25:391-405
  67. 67. Yang R, Argimon S, Li Y, Gu H, Zhou X, Caufield PW. Determining the genetic diversity of lactobacilli from the oral cavity. Journal of Microbiological Methods. 2010;82(2):163-169
  68. 68. ACR T, Kent RL Jr, Lif Holgerson P, Hughes CV, Loo CY, Kanasi E, et al. Microbiota of severe early childhood caries before and after therapy. Journal of Dental Research. 2011;90(11):1298-1305
  69. 69. Alcaraz LD, Belda-Ferre P, Cabrera-Rubio R, Romero H, Simón-Soro Á, Pignatelli M, et al. Identifying healthy oral microbiome through metagenomics. Clinical Microbiology and Infection. 2012;18(4):54-57
  70. 70. Belda-Ferre P, Alcaraz LD, Cabrera-Rubio R, Romero H, Simon-Soro A, Pignatelli M, et al. The oral metagenome in health and disease. The ISME Journal. 2012;6:46-56
  71. 71. Kawamura Y, Kamiya Y. Metagenomic analysis permit- ting identification of the minority bacterial populations in the oral microbiota. J. Oral Biosci. 2012;54:132-137
  72. 72. Pushalkar S, Ji X, Li Y, Estilo C, Yegnanarayana R, Singh B, et al. Comparison of oral microbiota in tumor and non-tumor tissues of patients with oral squamous cell carcinoma. BMC Microbiology. 2012;12:144
  73. 73. Reyes CPA, Dalmacio LMM. Bacterial diversity in the saliva and plaque of caries free and caries-active Filipino adults. Philippine Journal of Science. 2012;141(2):217-227
  74. 74. Yang F, Zeng X, Ning K, Liu KL, Wang W, Chen J, et al. Saliva microbiomes distinguish caries-active from healthy human populations. The ISME Journal. 2012;6(1):1-10
  75. 75. Benitez-Paez A, Belda-Ferre P, Simon-Soro A, Mira A. Microbiota diversity and gene expression dynamics in human oral biofilms. BMC Genomics. 2014;15:311
  76. 76. Mason MR, Preshaw PM, Nagaraja HN, Dabdoub SM, Rahman A, Kumar PS. He subgingival microbiome of clinically healthy current and never smokers. The ISME Journal. 2015;9(10):268-272
  77. 77. Schmidt BL, Kuczynski J, Bhattacharya A, Huey B, Corby PM, Queiroz ELS, et al. Changes in abundance or oral microbiota associated with oral cancer. PLoS One. 2014;9(8):e106297
  78. 78. Banas JA, Zhu M, Dawson DV, Cao H, Levy SM. PCR-based identification of oral streptococcal species. Int J Dent. 2016;2016:3465163
  79. 79. Dame-Teixeira N, Parolo CCF, Maltz M, Tugnait A, Devine D, and Do T. 2016. Actinomyces spp. gene expression in root canal lesions. J Oral Microbiol 8:10.3402.
  80. 80. Fourie NH, Wang D, Abey SK, Sherwin LB, Joseph PV, Rahim-Wil- liams B, Ferguson EG, and Henderson WA. The microbiome of the oral mucosa in irritable bowel syndrome. Gut Microbes. 2016;7:286-301
  81. 81. Al-hebshi NN, Nasher AT, Maryoud MY, Homeida HE, Chen T, Idris AM, et al. Inflammatory bacteriome featuring Fusobacterium nucleatum and Pseudomonas aeruginosa identified in association with oral squamous cell carcinoma. Scientific Reports. 2017;7:1834
  82. 82. Ganesan SM, Joshi V, Fellows M, Dabdoub SM, Nagaraja HN. A tale of two risks: Smoking diabetes and the subgingival microbiome. The ISME Journal. 2017;11:2075-2089
  83. 83. Lassalle F, Spagnoletti M, Fumagalli M, Shaw L, Dyble M, Walker C, et al. Oral microbiomes from hunter-gatherers and traditional farmers reveal shifts in commensal balance and pathogen load linked to diet. Mol Ecol. 2018 Jan;27(1):182-195. DOI: 10.1111/mec.14435
  84. 84. Mashima I, Theodorea CF, Thaweboon B, Thaweboon S, Scannapieco FA, Nakazawa F. Exploring the salivary microbiome of children stratified by the oral hygiene index. PLoS One. 2017;12(9):e0185274
  85. 85. Si J, Lee C, Ko G. Oral microbiota: Microbial biomarkers of metabolic syndrome independent of host genetic factors. Frontiers in Cellular and Infection Microbiology. 2017;7:516
  86. 86. Chen WP, Chang SH, Tang CY, Liou ML, Tsai SJJ, Lin YL. Composition analysis and feature selection of the oral microbiota associated with periodontal disease. BioMed Research International. 2018;2018:3130607
  87. 87. Thurnheer T, Belibasakis GN. Streptococus oralis maintains homeostasis in oral biofilms by antagonizing the cariogenic pathogen Streptococus mutans. Molecular Oral Microbiology. 2018;33(3):234-239
  88. 88. Carda-Diéguez M, Bravo-González LA, Morata IM, Vicente A, Mira A. High-throughput DNA sequencing of microbiota at interproximal sites. Journal of Oral Microbiology. 2020;12(1):1687397
  89. 89. Jiang Q, Liu J, Chen L, Gan N, Yang D. The Oral Microbiome in the Elderly With Dental Caries and Health. Front Cell Infect Microbiol. 2019;8:442. Published 2019 Jan 4. doi:10.3389/fcimb.2018.00442
  90. 90. Raju SC, Lagström S, Ellonen P, de Vos WM, Eriksson JG, Weiderpass E, et al. Gender-specific associations between saliva microbiota and body size. Frontiers in Microbiology. 2019;10:767
  91. 91. Thurnheer T, Karygianni L, Flury M, and Belibasakis GN. 2019. Fusobacterium species and subspecies differentially affect the composition and architecture of supra- and subgingival biofilms models. Front Microbioldoi.org/10.3389/fmicb.2019.01716
  92. 92. Burcham ZM, Garneau NL, Comstock SS, Tucker RM, Knight R, Metcalf JL and Genetics of Taste Lab Citizen Scientists. 2020. Patterns of Oral microbiota diversity in adults and children: A crowdsourced population study. Scientific Reports 10:2133.
  93. 93. Kori J, Saleem F, Ullah S, Azim K. Characterization of Oral bacteriome dysbiosis in type 2 diabetic patients. In: medRxiv. 2020
  94. 94. Handelsman J. Metagenomics: Application of genomics to uncultured microorganisms. Microbiology and Molecular Biology Reviews. 2002;68:669-685
  95. 95. Sizova M, Hohmann T, Hazen A, Paser BJ, Halem SR, Murphy CM, et al. New approaches for isolation of previously uncultivated oral bacteria. Applied and Environmental Microbiology. 2012;78:194-203
  96. 96. Asikainen S, Karched M, Rogers A. Molecular Techniques in Oral Microbial Taxonomy Identifications and Typing. Norfolk, UK: Caister Academic Press; 2008
  97. 97. Zaura E. Next-generation sequencing approaches to understanding the oral microbiome. Advances in Dental Research. 2012;24:81-85
  98. 98. Soro V, Dutton LC, Sprague SV, Nobbs AH, Ireland AJ, Sandy JR, et al. Axenic culture of a can- didate division TM7 bacterium from the human oral cavity and biofilm interactions with other oral bacteria. Applied and Environmental Microbiology. 2014;80:6480-6489
  99. 99. Wade WG. Unculturable bacteria—The uncharacterized organisms that cause oral infections. Journal of the Royal Society of Medicine. 2002;95:81-83
  100. 100. Jenkinson HF. Beyond the oral microbiome. Environmental Microbiology. 2011;13:3077-3087
  101. 101. Siqueira JF Jr, Rôças IN. As-yet-uncultivated oral bacteria: Breadth and association with oral and extra-oral diseases. Journal of Oral Microbiology. 2013;5:1-14
  102. 102. Benn A, Heng N, Broadbent JM, Thomson WM. 2018. Studying the human oral microbiome: Challenges and the evolution of solutions. Australian Dental Journal. 2018;63:14-24
  103. 103. Anderson IC, Cairney JW. Diversity and ecology of soil fungal communities: Increased understanding through the application of molecular techniques. Environmental Microbiology. 2004;6:769-779
  104. 104. Nishigaki K, Naimuddin M, Hamano K. Genome profiling: A realistic solution for genotype-based identification of species. Journal of Biochemistry. 2000;128:107-112
  105. 105. Socransky SS, Smith C, Martin L, Paster BJ, Dewhirst FE, Levin AE. “Checkerboard” DNA-DNA hybridization. BioTechniques. 1994;17:788-792
  106. 106. Schena M, Shalon D, Davis RW, and Brown PO. 1995. Quantitative monitoring of gene expression patterns with a complementary DNA microarray. Science270:467–470.
  107. 107. Chung SK. Oral metagenomic analysis techniques. J Dent Hyg Sci. 2019;19(2):86-95
  108. 108. Chen K, Neimark H, Rumore P, Steinman CR. Broad-range DNA probes for detecting and amplifying eubacterial nucleic acids. FEMS Microbiology Letters. 1989;57:19-24
  109. 109. Relman DA. The search for unrecognized pathogens. Science. 1999;284:1308-1310
  110. 110. Yang B, Wang Y, Qian PY. Sensitivity and correlation of hypervariable regions in 16S rRNA genes in phylogenetic analysis. BMC Bioinformatics. 2016;17:135
  111. 111. Chakravorty S, Helb D, Burday M, Connell N, Alland D. A detailed analysis of 16S ribosomal RNA gene segments for the diagnosis of pathogenic bacteria. Journal of Microbiological Methods. 2007;69:330-339
  112. 112. Kumar PS, Brooker MR, Dowd SE, Camerlengo T. Target region selection is a critical determinant of community fingerprints generated by 16S pyrosequencing. PLoS One. 2011;6:e20956
  113. 113. Kawamura Y, Fukunaga H, Hirose K, Ezaki T. Basic study of the LCR-MTB system. Clin Microbiol. 1998;25:873-874
  114. 114. Kuwamura Y, Kamiya Y. Metagenomic analysis permitting identification of the minority bacterial populations in the oral microbiota. Journal of Oral Biosciences. 2012;54(3):132-137
  115. 115. Avila M, Ojcius DM, Yilmaz O. The oral microbiota: Living with a permanent guest. DNA and Cell Biology. 2009;28(8):405-411
  116. 116. Elahi E, Ronaghi M. Pyrosequencing: A tool for DNA sequencing analysis. Methods in Molecular Biology. 2004;255:211-219
  117. 117. Ames NJ, Ranucci A, Moriyama B, Wallen GR. The human microbiome and understanding the 16S rRNA gene in translational nursing science. Nursing Research. 2017;66(2):184-197
  118. 118. Hasan NA, Young BA, Minard-Smith AT, Saeed K, Li H, Heizer EM, et al. Microbial community profiling of human saliva using shotgun metagenomic sequencing. PLoS One. 2014;9(5):e97699
  119. 119. Yu G, Phillips S, Gail MH, Goedert JJ, Humphrys M, Ravel J, et al. Evaluation of buccal cell samples for studies of oral microbiota. Cancer Epidemiology, Biomarkers & Prevention. 2017;26:249-253
  120. 120. Clemente JC, Pehrsson EC, Blaser MJ, Sandhu K, Gao Z, Wang B, Magris M, Hidalgo G, Contreras M, Noya-Alarcon O, Lander O, McDonald J, Cox M, Walter J, Oh PL, Ruiz JF, Rodriguez S, Shen N, Song SJ, Metcalf J, Knight R, Dantas G, Dominguez-Bello MG. 2015. The microbiome of uncontacted Amerindians. Sci Adv. 2015;1:e1500183.
  121. 121. Lim MY, Cho Y, Rho M. Diverse distribution of Resistomes in the human and environmental microbiomes. Current Genomics. 2018;19(8):701-711
  122. 122. Choi Y-H, Kosaka T, Ojima M, Sekini S, Kokubo Y, Watanabe M, et al. Relationship between the burden of major periodontal bacteria and serum lipid profile in a cross-sectional Japanese study. BMC Oral Health. 2018;18(1):77
  123. 123. Topcuoglu N, Kulekci G. 16S rRNA based microarray analysis of ten periodontal bacteria in patients with different forms of periodontitis. Anaerobe, Volume 35. Part A. 2015;2015:35-40
  124. 124. Zheng H, Xu L, Wang Z, Li L, Zhang J, Zhang Q, et al. Subgingival microbiome in patients with healthy and ailing dental implants. Sci Rep. 2015 Jun 16;5:10948
  125. 125. Hu X, Zhang Q, Hua H, Chen F. Changes in the salivary microbiota of oral leukoplakia and oral cancer. Oral Oncol. 2016;56:6-8
  126. 126. Amer A, Galvin S, Healy CM, Moran GP. The microbiome of potentially malignant oral leukoplakia exhibits enrichment for Fusobacterium, Leptotrichia, Campylobacter, and Rothia species. Front Microbiol. 2017;8:2391-2400
  127. 127. Wang Y, Zhang J, Chen X, et al. Profiling of oral microbiota in early childhood caries using single-molecule real-time sequencing. Front Microbiol. 2017;8:2244
  128. 128. Agnello M, Cen L, Tran NC, Shi W, Mclean JS, He X. Arginine improves pH homeostasis via metabolism and microbiome modulation. J. Dent. Res. 2017;96:924-930
  129. 129. Said HS, Suda W, Nakagome S, Chinen H, Oshima K, Kim S, Kimura R, Iraha A, Ishida H, Fujita J, Mano S, Morita H, Dohi T. Oota H, and Hattori M. 2014. Dysbiosis of salivary microbiota in inflammatory bowel disease and its association with oral immunological biomarkers. DNA Research 21:15–25.
  130. 130. Mager DL, Haffajee AD, Devlin PM, Norris CM, Posner MR, Goodson JM. The salivary microbiota as a diagnostic indicator of oral cancer: a descriptive, non-randomized study of cancer-free and oral squamous cell carcinoma subjects. J Transl Med. 2005;3:27
  131. 131. Mager DL, Haffajee AD, Devlin PM, Norris CM, Posner MR, Goodson JM. The salivary microbiota as a diagnostic indicator of oral cancer: a descriptive, non-randomized study of cancer-free and oral squamous cell carcinoma subjects. J Transl Med. 2005;3:27
  132. 132. Pushalkar S, Mane SP, Ji X, et al. Microbial Diversity in Saliva of Oral Squamous Cell carcinoma. FEMS Immunol Med Microbiol. 2011;61(3):269-277
  133. 133. Al-Hebshi NN, Nasher AT, Maryoud MY, et al. Inflammatory bacteriome featuring Fusobacetrium nucleatum and pseudomonas aeruginosa identified in association with oral squamous cell carcinoma. Sci Rep. 2017;7(1):1834
  134. 134. Lee WH, Chen H-M, Yang S-F, Liang C, Peng C-Y, Lin F-M, et al. Bacterial alterations in salivary microbiota and their association in oral cancer. Sci. Rep. 2017;7
  135. 135. Torres PJ, Fletcher EM, Gibbons SM, Bouvet M, Doran KS, Kelley ST. Characterization of the salivary microbiome in patients with pancreatic cancer. PerrJ 2015; 3e1373
  136. 136. Fan X, Alekseyenko AV, Wu J, Peters BA, Jacobs EJ, Gapstur SM, et al. Human oral microbiome and prospective risk for pancre- atic cancer: a population-based nested case-control study. Gut. 2016;67(1):120-127
  137. 137. Olson SH, Satagopan J, Xu Y, et al. The oral microbiota in patients with pancreatic cancer, patients with IPMNs and controls: a pilot study. Cancer Causes Control. 2017;28(9):959-969
  138. 138. Lu H, Ren Z, Li A. Tongue coating microbiome data distinguish patients with pancreatic head cancer from health controls. Journal of Oral Microbiology. 2019;11(1):1563409
  139. 139. Katz J, Onate MD, Pauley KM, Bhattacharyya I, Cha S. Presence of Porphyromonas gingivalis in gingival squamous cell carcinoma. Int J Oral Sci. 2011;3(4):209-215
  140. 140. Guerrero-Preston R, White JR, Godoy-Vitorino F, et al. High-resolution microbiome profiling uncovers Fusobacterium nucleatum, lactobacillus gasseri/johnsonii and Lactobacillus vaginalis associated to oral and oropharyngeal cancer in saliva from HPV-positive and HPOV negative patients treated with surgery and chemo-radiation. Oncotarget. 2017;8(67):110931-110948
  141. 141. Decsi G, Soki J, Pap B, Dobra G, Harmati M, Kormondi S, et al. Chicken or the egg: Microbial alterations in biopsy samples of patients with oral potentially malignant disorders. Pathol Oncol Res. 2018. [PMID: 30054809]
  142. 142. Scalas D, Roana J, Boffano P, et al. Bacteriological findings in radicuylar cyst and keratocystic odontogenic tumuor fludis from asymptomatic patients. Arch Oral Biol. 2013;58(11):1578-1583
  143. 143. Flemer B, Lynch DB, Brown JM, et al. Tumour-associated and non-tumour-associated microbiota in colorectal cancer. Gut. 2017;66(4):633-643
  144. 144. Peters BA, Wu J, Pei Z, Yang L, Purdue MP, Freedman ND, et al. Oral Microbiome composition reflects prospective risk for esophageal cancers. Cancer Res. 2017;77(23):6777-6787
  145. 145. Chen, H., Liu, Y., Zhang, M., Wang, G., Qi, Z., Bridgewater, L., ... Pang, X. (2015). A Filifactor alocis-centered co-occurrence group associates with periodontitis across different oral habitats. Scientific Reports, 5.
  146. 146. Wu J, Xu S, Xiang C, et al. Tongue Coating Microbiota Community and Risk Effect on Gastric Cancer. J Cancer. 2018;9(21):4039-4048
  147. 147. Mukherjee PK, Wang H, Retuerto M, et al. Bacteriome and mycobiome associatoons in oral tongue cancer. Oncotarget. 2017;8(57):97273-97289
  148. 148. Kamer AR, Craig RG, Pirraglia E, Dasanayake AP, Norman RG, Boylan RJ, et al. TNF-alpha and antibodies to periodontal bacteria discriminate between Alzheimer’s disease patients and normal subjects. J Neuroimmunol. 2009;216:92-97
  149. 149. Kamer AR, Craig RG, Pirraglia E, Dasanayake AP, Norman RG, Boylan RJ, et al. TNF-alpha and antibodies to periodontal bacteria discriminate between Alzheimer’s disease patients and normal subjects. J Neuroimmunol. 2009;216:92-97
  150. 150. Casarin RC, Barbagallo A, Meulman T, Santos VR, Sallum EA, Nociti FH, et al. Subgingival biodiversity in subjects with uncontrolled type-2 diabetes and chronic periodontitis. J Periodontal Res. 2013;48(1):30-36
  151. 151. Madianos PN, Bobetsis YA, Offenbacher S. Adverse pregnancy outcomes (APOs) and periodontal disease: Patho- genic mechanisms. Journal of Clinical Periodontology. 2013;40(14):S170-S180
  152. 152. Han YW, Fardini Y, Chen C, Iacampo KG, Peraino VA, Shamonki JM, et al. Term stillbirth caused by oral Fusobacterium nucleatum. Obstetrics and Gynecology. 2010;115:442-445
  153. 153. Zhang X, Zhang D, Jia H, Feng Q, Wang D, Liang D, et al. The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment. Nature Medicine. 2015;21(8):895-905
  154. 154. Dang AT, Cotton S, Sankaran-Walters S, Li CS, Lee CY, Dandekar S, et al. Evidence of an increased pathogenic footprint in the lingual micro- biome of untreated HIV infected patients. BMC Microbiol. 2012;12:153
  155. 155. Koren O, Spor A, Felin J, Fak F, Stombaugh J, Tremaroli V, et al. Human oral, gut, and plaque mi- crobiota in patients with atherosclerosis. Proc. Natl. Acad. Sci. U. S. A. 2011;108(Suppl. 1):4592-4598
  156. 156. Rôças IN, Alves FRF, Rachid CTCC, Lima KC, Assunção IV, Gomes PN, Siqueira JFJr. 2016. Microbiome of deep dentinal caries lesions in teeth with symptomatic irreversible pulpitis. PLoS One 2016;11:e0154653.
  157. 157. Takeshita T, Matsuo K, Furuta M, Shibata Y, Fukami K, Shimazaki Y, et al. Distinct composition of the oral indigenous microbiota in south Korean and Japanese adults. Scientific Reports. 2014;4:6990
  158. 158. Zhou J, Jiang N, Wang S, Hu X, Jiao K, He X, et al. Exploration of human salivary microbiomes-insights into the novel characteristics of microbial community structure in caries and caries-free subjects. PLoS One. 2016;11(1):e0147039
  159. 159. Han YW, Wang X. Mobile microbiome: Oral bacteria in extra-oral infections and inflammation. Journal of Dental Research. 2013;92:485-491
  160. 160. Gao L, Xu T, Huang G, Jiang S, Gu Y, Chen F. Oral microbiomes: More and more importance in oral cavity and whole body. Protein & Cell. 2018;9(5):488-500
  161. 161. Huang X, Palmer SR, Ahn SJ, Richards VP, Williams ML, Nascimento MM, et al. A highly arginolytic streptococcus species that potently antagonizes Streptococcus mutans. Applied and Environmental Microbiology. 2016;82:2187-2201
  162. 162. Curtis MA. Periodontal microbiology - the lid’s off the box again. Journal of Dental Research. 2014;93:840-842
  163. 163. Yakob M, Fuentes L, Wang MB, Abemayor E, Wong DTW. Salivary biomarkers for detection of oral squamous cell carcinoma: Current state and recent advances. Curr Oral Health Rep. 2014;1:133-141
  164. 164. Marsh PD. In sickness and in health-what does the oral microbiome mean to us? An ecological perspective. Advances in Dental Research. 2018;29(1):60-65
  165. 165. Johansson A, Claesson R, Höglund Åberg C, Haubek D, Lindholm M, Jasim S, et al. Genetic profiling of Aggregatibacter actinomycetemcomitans serotype B isolated from periodontitis patients living in Sweden. Pathogens. 2019; 8(3):153
  166. 166. Esberg A, Sheng N, Mårell L, Claesson R, Persson K, Borén T, et al. Streptococcus Mutans adhesin biotypes that match and predict individual caries development. EBio Med. 2017;24:205-215
  167. 167. Yamanaka W, Takeshita T, Shibata Y, Matsuo K, Eshima N, Yokoyama T, et al. Compositional stability of a salivary bacterial population against supragingival microbiota shift following periodontal therapy. PLoS One. 2012;7(8):e42806
  168. 168. Nathan C. Antibiotics at the crossroads. Nature. 2004;431(7011):899-902
  169. 169. Ready D, Lancaster H, Qureshi F, Bedi R, Mullany P, Wilson M. Effect of amoxicillin use on oral microbiota in young children. Antimicrobial Agents and Chemotherapy. 2004;48(8):2883-2887
  170. 170. Guarner F, Perdigon G, Corthier G, Salminen S, Koletzko B, Morelli L. Should yoghurt cultures be considered probiotic? The British Journal of Nutrition. 2005;93(6):783-786
  171. 171. Teughels W, Loozen G, Quirynen M. Do probiotics offer opportunities to manipulate the periodontal oral microbiota? Journal of Clinical Periodontology. 2011;38:159-177
  172. 172. Lingam L, Ramesh T, Lavanya R. Bacteria in oral health-probiotics and prebiotics a review. Int. J. Biol. Med. Res. 2011;2(4):1226-1233
  173. 173. Meurman JH, Stamatova I. Probiotics: Contributions to oral health. Oral Diseases. 2007;13(5):443-451
  174. 174. Busscher H, Mulder A, Van der Mei H. In vitro adhesion to enamel and in vivo colonization of tooth surfaces by lactobacilli from a bio–yoghurt. Caries Research. 1999;33(5):403-404
  175. 175. Näse L, Hatakka K, Savilahti E, Saxelin M, Pönkä A, Poussa T, et al. Effect of of long–term consumption of a probiotic bacterium, lactobacillus rhamnosus GG, in milk on dental caries and caries risk in children. Caries Research. 2001;35(6):412-420
  176. 176. Caglar E, Sandalli N, Twetman S, Kavaloglu S, Ergeneli S, Selvi S. Effect of yogurt with bifidobacterium DN-173 010 on salivary mutans streptococci and lactobacilli in young adults. Acta Odontologica Scandinavica. 2005;63(6):317-320
  177. 177. Caglar E, Kavaloglu S, Cildir S, Ergeneli S, Sandalli N, Twetman S. Salivary mutans streptococci and lactobacilli levels after ingestion of the probiotic bacterium lactobacillus reuteri ATCC 55730 by straws or tablets. Acta Odontologica Scandinavica. 2006;64(5):314-318
  178. 178. Cildir SK, Germec D, Sandalli N, Ozdemir FI, Arun T, Twetman S, et al. Reduction of salivary mutans streptococci in orthodontic patients during daily consumption of yoghurt containing probiotic bacteria. European Journal of Orthodontics. 2009;31(4):407-411
  179. 179. Holz C, Alexander C, Balcke C, More M, Auinger A, Bauer M, et al. Lactobacillus paracasei DSMZ16671 reduces mutans streptococci: A short-term pilot study. Probiotics Antimicrob. Proteins. 2013;5:259-263
  180. 180. Ohshima T, Kojima Y, Seneviratne CJ, Maeda N. Therapeutic application of synbiotics, a fusion of probiotics and prebiotics, and biogenics as a new concept for oral Candida infections: A mini review. Front Microbiol doi. 2016. DOI: 10.3389/fmicb.2016.00010
  181. 181. Zarco MF, Vess TJ, Ginsburg GS. The oral microbiome in health and disease and the potential impact on personalized dental medicine. Oral Diseases. 2012;18(2):109-120
  182. 182. Tagg JR, Dierksen KP. Bacterial replacement therapy: Adapting’ germ warfare’ to infection prevention. Trends in Biotechnology. 2003;21(5):217-223
  183. 183. Ren Z et al. Dual-targeting approach degrades biofilm matrix and enhances bacterial killing. J. Dent. Res. 2019;98:322-330
  184. 184. Paisano AF, Spira B, Cai S, Bombana AC. In vitro antimicrobial effect bacteriophages on human dentin infected with enterococcus faecalis ATCC 29212. Oral Microbiol. Immunol. 2004;19(5):327-330
  185. 185. Benergossi J, Calixto G, Fonseca-Santos B, Aida KL, Negrini TC, Duque C, et al. Highlights in peptide nanoparticle carriers intended to oral diseases. Current Topics in Medicinal Chemistry. 2015;15(4):345-355
  186. 186. Hearnden V, Sankar V, Hull K, Juras DV, Greenberg M, Kerr AR, et al. New developments and op- portunities in oral mucosal drug delivery for local and systemic disease. Advanced Drug Delivery Reviews. 2012;64(1):16-28
  187. 187. Joshi D, Garg T, Goyal AK, Rath G. Advanced drug delivery approaches against periodontitis. Drug Delivery. 2016;23(2):363-377
  188. 188. Rosier BT, Marsh PB, Mira A. Resilience of the Oral microbiota in health: Mechanisms that prevent dysbiosis. Journal of Dental Research. 2018;97(4):371-380
  189. 189. Department of Health Philippines. National Monitoring & Evaluation Dental Survey (2011): Final Report. Philippines: Manila; 2013
  190. 190. Department of Health Philippines. 2007. Administrative Order 2007–0007 Guidelines in Implementation of Oral Health Program for Public Health Services.

Written By

Marilen P. Balolong and Michael Antonio F. Mendoza

Submitted: 26 May 2020 Reviewed: 26 October 2020 Published: 08 February 2021