Download as pdf or txt
Download as pdf or txt
You are on page 1of 4

Journal of Oral Biosciences 54 (2012) 128131

Contents lists available at SciVerse ScienceDirect

Journal of Oral Biosciences


journal homepage: www.elsevier.com/locate/job

Review

Evaluation of salivary microbiota from an ecological perspective


Toru Takeshita n, Yoshihisa Yamashita
Section of Preventive and Public Health Dentistry, Division of Oral Health, Development and Growth, Kyushu University Faculty of Dental Science, Fukuoka 812-8582, Japan

a r t i c l e i n f o a b s t r a c t

Article history: A variety of bacteria densely colonize the human oral cavity not as individuals, but as members of an
Received 13 January 2012 indigenous microbiota that exhibit extensive intracellular interactions. To prevent the onset of oral
Received in revised form diseases caused by the members of this microbiota, regulation of the total oral microbiota, including
26 March 2012
the surrounding bacterial environment, is required. In this review, we highlight current knowledge on
Accepted 4 April 2012
Available online 9 August 2012
the global composition of the salivary bacterial population associated with oral conditions through a
molecular ecological approach using the 16S rRNA gene. The salivary bacterial populations of Japanese
Keywords: subjects were commonly dominated by bacterial genera such as Streptococcus, Prevotella, and Neisseria,
Microbiota and their relative abundances differed according to the specic periodontal condition of the patient.
16S rRNA
This suggests that manipulation of the predominant oral microbiota may assist in the maintenance of
T-RFLP
periodontal health. A broad view of the relationship of oral health with oral microbes would provide
Saliva
novel insights into oral health and disease.
& 2012 Japanese Association for Oral Biology. Published by Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128
2. Molecular ecological approach using the 16S ribosomal RNA gene. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129
3. T-RFLP analysis of the bacterial community in saliva . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129
4. Further analysis of the salivary microbiota using the clone library method . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 130
5. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 131
Conict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 131
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 131

1. Introduction of oral environmental changes, resulting in caries [5,6]. However,


bacteriological etiologies of these diseases, especially periodontitis,
Two major oral diseases, namely, dental caries and period- have not yet been elucidated.
ontitis are infectious diseases that are caused by bacteria. Several Numerous and diverse microorganisms colonize the human oral
specic microbes are assumed to be causal agents, and their cavity as commensals. The number of bacterial species identied
etiologies have been extensively pursued. Currently, little doubt exceeds 700 [7]. These microorganisms regulate each other and
exists that mutans streptococci are associated with the initiation coexist within the microbial community, i.e., the indigenous micro-
of dental caries [13], and Porphyromonas gingivalis, Tannerella biota. Oral pathogenic bacteria are also members of this complex
forsythensis, Treponema denticola, and a complex formed by these microbial community, and therefore, their growth and the expres-
bacteria are prime suspects in periodontitis [4]. However, the sion of virulence factors are most likely to be affected by synergistic
presence of these organisms is not required for the onset of oral or antagonistic interactions with other indigenous bacteria.
diseases. In the study of dental caries, the ecological plaque The surrounding bacterial populations, even those with no direct
hypothesis, an excellent hypothesis suggesting the etiology of virulence factors, may thus be involved in the onset of oral disease.
this disease, recently emerged; this hypothesis proposes that Recent advances in molecular ecological techniques have allowed
dental plaque microbiota shift to an aciduric community because comprehensive surveys of complex bacterial communities, and the
overall structure of bacterial communities formed within various oral
sites has been revealed [811]. In particular, dental plaque micro-
n
Corresponding author. biota associated with dental caries and periodontitis have been
E-mail address: taketooo@dent.kyushu-u.ac.jp (T. Takeshita). investigated in detail, and many novel putative causative agents,

1349-0079/$ - see front matter & 2012 Japanese Association for Oral Biology. Published by Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.job.2012.04.003
T. Takeshita, Y. Yamashita / Journal of Oral Biosciences 54 (2012) 128131 129

including not-yet-cultivated microorganisms, have been identied their size, and their uorescent intensities reect the abundance of
[1216]. that bacterium in the microbiota. The peak pattern represents an
On the other hand, dental plaque microbiota are continuously overview of the microbial community structure that can be easily
bathed in saliva and are therefore always in contact with the compared with other samples, although additional work is required
bacterial population in saliva. Although the salivary bacterial for phylogenetic interpretation of a peak pattern. This method is
population is a mixture of bacterial communities that exist at especially powerful for monitoring shifts in microbiota and has been
various sites in the oral cavity, its composition is more similar to adopted for characterizing these oral microbiota shifts [23].
the microbiota on mucosal surfaces, which may act as a reservoir Both sequencing and ngerprinting have their advantages and
for dental pathogens, compared to dental plaque [17]. We hope disadvantages, and thus, a combination of these 2 methods
that the observation of salivary bacterial populations may provide is required to compare large numbers of complex bacterial
further insights into the involvement of oral microbiota in the community samples. To characterize the relationship between
oral environment, affecting the onset of oral diseases. salivary bacterial population structure and oral health, we primarily
In this review, we introduce our progress regarding the utilized the T-RFLP method. Our efforts to improve fragment size
relationship between the overall structure of the salivary bacterial accuracy [24] have allowed for more accurate predictions of
population and oral health and disease. bacterial compositions from a T-RFLP peak pattern, and the applica-
tion of this method has been specialized for the analysis of oral
microbiota [25]. The microbiota structures characterized by T-RFLP
2. Molecular ecological approach using the 16S ribosomal analysis were conrmed using a clone library analysis of some
RNA gene representative samples.

In the past, comprehensive characterization and comparison of


multiple complex bacterial communities, such as dental plaque 3. T-RFLP analysis of the bacterial community in saliva
and saliva, have been regarded as unrealistic because conven-
tional methods rely on the cultivation of bacteria and are To explore the relationship between the salivary bacterial
laborious, time-consuming, and expensive. In addition, noncul- population and oral health, we examined the oral condition of
turable bacteria can be missed. A molecular ecological approach 200 Japanese subjects, ages 1540 years, and collected their saliva
using DNA extracted from the microbial community overcomes [26]. The bacterial composition and communities in the saliva of
these limitations. Use of the 16S ribosomal RNA (rRNA) gene in these 200 subjects was investigated using T-RFLP analysis of the
combination with polymerase chain reaction (PCR) has become 16S rRNA gene, and data were displayed as peak patterns.
common in analyses of bacterial communities. The 16S rRNA gene We used forward and reverse primers labeled with the uorescent
(total length: approximately 1500 bp) is present in almost all dyes 6-carboxyuorescein (6-FAM) and hexachloro uorescein
bacterial species. This gene has remained relatively conserved (HEX), respectively; thus 2 peak patterns per sample were
throughout evolution, but contains 9 variable regions that can be obtained in each electrophoretic run. T-RFLP proles of the 200
utilized for classifying bacteria at the species level [18]. PCR using subjects contained 110 distinct peaks, 69 in the 6-FAM proles
a primer set that targets a conserved region of the 16S rRNA gene (F1F69) and 41 in the HEX proles (R1R41). No peaks were
allows us to collect genes from nearly all members of the bacterial specically detected in subjects with many decayed teeth, no
community. caries, severe periodontitis, and healthy gingiva. In contrast, large
Two major approaches are used to analyze PCR amplicon peaks were detected at the same position in almost all T-RFLP
sequence information: sequencing and ngerprinting. In clone proles, suggesting that the dominant indigenous bacteria in
library analysis (the sequencing approach), amplicons are cloned saliva were common to all subjects. In addition, their relative
into a plasmid vector, and the resultant plasmids are transformed abundances among subjects differed considerably.
into Escherichia coli. The inserts are sequenced individually and To correlate the T-RFLP peak patterns with oral health condi-
compared to sequences of known origin deposited in a public tions, we classied subjects based on their T-RFLP proles and
database. Members of bacterial communities can be identied compared the oral condition of subjects in each group. Three
individually. Using this method, previous studies have documen- major patterns were identied from the 200 proles using the
ted the overall composition of various oral bacterial communities, partitioning around medoids analysis (Fig. 1). The relative abun-
such as supragingival plaques of subjects with periodontitis and dances of dominant peaks were different in each of the 3 clusters.
periodontally healthy subjects [9], tongue coatings of subjects For example, peaks F19 and F33 were characteristically more
with and without halitosis [10], dentoalveolar abscesses [11], and predominant in the 6-FAM T-RFLP proles of cluster I compared
root canal microbiota associated with treatment failures [19].
In addition, a database limited to oral bacteria was constructed [20],
and it can be used to facilitate analysis of the oral bacterial
community.
However, while the cloning and sequencing method does generate
accurate phylogenic information, is somewhat laborious for identi-
cation of the overall microbiota structure because of the need to
sequence many clones. Fingerprinting approaches, such as terminal
restriction fragment length polymorphism (T-RFLP) [21] and denatur-
ing gradient gel electrophoresis [22], are more convenient and allow
rapid assessment and comparison of a large number of bacterial
community samples. For example, in T-RFLP analysis, 16S rRNA genes
are amplied using uorescent-labeled primers, and the amplicons
are digested with a restriction enzyme. The resultant fragments are Fig. 1. Peak patterns of 6-carboxyuorescein-labeled terminal restriction frag-
ment length polymorphisms showing the bacterial composition of the saliva of
separated by capillary electrophoresis, and only the uorescent- 200 subjects displayed as a 3-dimensional plot. Peaks (x-axis), subjects sorted into
labeled 5-terminal-labeled fragments are detected. Labeled frag- each cluster (41, 74, and 85 subjects in clusters I, II, and III, respectively; y-axis),
ments from differing bacterial origins were discriminated based on and area proportion of each peak (z-axis) are shown.
130 T. Takeshita, Y. Yamashita / Journal of Oral Biosciences 54 (2012) 128131

Table 1
Comparison of oral conditions among 3 clusters of subjects classied based on terminal restriction fragment length polymorphism proles.

Parameters Cluster I Cluster II Cluster III Statistical


(n 41) (n 74) (n 85) difference

Teeth condition
Number of missing teeth (mean 7 SD) 0.27 0.6 0.4 7 1.0 0.27 0.7 NSa
Number of DMFT (mean 7 SD) 9.47 6.4 10.1 7 7.1 7.87 5.4 NSa
Periodontal condition
Percentage of sites bleeding on probing (mean 7SD) 27.6 7 18.1 18.9 7 19.8 14.67 13.5 I 4II, I 4IIIa
Percentage of sites with periodontal pockets 10.17 15.4 9.0 7 15.1 3.77 6.4 I 4III, II4IIIa
(mean 7 SD)
(pocket depth 44 mm)
Number of subjects without periodontal pockets (%) 10 (24.3) 36 (48.6) 55 (64.7) P o0.001b
Number of subjects with periodontal pockets 420% (%) 9 (21.9) 14 (18.9) 2 (2.3) P o0.001b

Abbreviations: DMFT, decayed, missing, lled teeth; SD, standard deviation; NS, not signicant.
a
Statistical differences were evaluated among all 3 clusters by the Steel-Dwass test. A P valueo 0.05 was considered statistically signicant.
b
Statistical differences were evaluated by Fishers exact test.

other clusters. Peaks with large loading in the direction of cluster II


were F47 and R32, and Streptococcus species such as Streptococcus
mitis and S. salivarius were assigned to these peaks. Peaks with
large loading in the direction of cluster III were F17, F65, R12,
and R24, which corresponded to Neisseria, Haemophilus, Aggrega-
tibacter, and Porphyromonas species, excluding P. gingivalis. All
peaks with large loading were detected in every subject with a
large peak area, and the candidates corresponding to each peak
were common bacteria in human saliva.

4. Further analysis of the salivary microbiota using the clone


library method

To conrm the bacterial diversity of the salivary microbiota,


we selected 1 subject from each cluster and investigated their
microbiota using clone library analysis. The T-RFLP proles of
these representatives corresponded to the cluster center, imply-
ing that their microbiota structures were the most typical in each
cluster. As expected, common oral bacteria such as S. salivarius,
P. melaninogenica, and Veillonella dispar predominated in the
saliva of all subjects, consistent with bacterial prediction based
Fig. 2. Principal components analysis biplot showing the relationships among 200
on the T-RFLP proles. In addition, the relative abundance of each
terminal restriction fragment length polymorphism proles classied into 3 clus-
ters (3 types of dots) and each peak (arrows). Only 12 peaks with large loadings on bacterial genus differed among subjects, also consistent with the
the rst 2 principal components of 110 peaks were selected and shown. Candidate T-RFLP proles (Fig. 3).
bacterial genera corresponding to peaks, as determined by fragment size, are The results of this investigation suggest that the global
indicated. composition of the oral indigenous microbiota reected gingival
health. A microbiota dominated by Prevotella and Veillonella was
with the other clusters. The gingival health conditions of subjects implicated in periodontal disease, while that dominated by
in these 3 clusters were signicantly different (Table 1). Clusters I Neisseria, Haemophilus, Aggregatibacter, and Porphyromonas was
and II had a higher percentage of sites in periodontal pockets associated with gingival health. This result is noteworthy because
greater than 4 mm, and cluster I most often contained sites that periodontitis is local inammation of the gingival tissue, which is
exhibited bleeding on probing. Signicant differences among the thought to be an infectious disease caused by pathogenic bacteria
clusters were also observed in a number of subjects without inhabiting the gingival crevice. Although periodontal pathogens
periodontal pockets and with periodontal pockets greater than are also detectable in saliva [27,28], it is difcult for saliva to
20%. No signicant differences were observed for other clinical access the inside of periodontal pockets compared with other oral
parameters, including dental caries history. surfaces. Most importantly, differences in microbiota structures
T-RFLP proles were further subjected to principal compo- were determined by the relative abundance of the dominant
nents analysis (PCA) to characterize the bacterial community indigenous bacteria; recognized pathogens, such as P. gingivalis,
structures of each cluster (Fig. 2). Subjects within each cluster were not included in the T-RFLP peaks characteristic of each
were localized differently in the PCA plot of the rst 2 compo- cluster. Both the local emergence of pathogens and structural
nents. Subjects in cluster I were localized in the lower left region, differences in the entire oral microbiota are associated with
and peaks F19, F33, R16, and R22 had large loading in this gingival disorders. These characteristic dominant indigenous
direction. Based on fragment size, Prevotella species, including bacteria may be nonparticipating observers that prefer the oral
Prevotella melaninogenica, and Veillonella species were selected environment during periodontitis or periodontal health. However,
from our database (containing 655 oral bacterial species) as numerous interactions occur among pathogenic and other indi-
corresponding to the combination of these peaks, suggesting that genous bacteria within the oral microbiota. This study likely
these bacteria were more predominant in cluster I than in the characterized the bacterial environment surrounding a specic
T. Takeshita, Y. Yamashita / Journal of Oral Biosciences 54 (2012) 128131 131

of monoclonal antibodies to eight serotypes of the mutans streptococci.


Caries Res 2006;40:614.
[3] Loesche WJ. Role of Streptococcus-mutans in human dental decay. Microbiol
Rev 1986;50:35380.
[4] Socransky SS, Haffajee AD, Cugini MA, Smith C, Kent Jr. RL. Microbial
complexes in subgingival plaque. J Clin Periodontol 1998;25:13444.
[5] Marsh PD. Dental plaque as a biolm and a microbial community
implications for health and disease. BMC Oral Health 2006;6(Suppl 1):S14.
[6] Takahashi N, Nyvad B. The role of bacteria in the caries process: ecological
perspectives. J Dent Res 2011;90:294303.
[7] Dewhirst FE, Chen T, Izard J, Paster BJ, Tanner AC, Yu WH, Lakshmanan A,
Wade WG. The human oral microbiome. J Bacteriol 2010;192:500217.
[8] Aas JA, Paster BJ, Stokes LN, Olsen I, Dewhirst FE. Dening the normal
bacterial ora of the oral cavity. J Clin Microbiol 2005;43:572132.
[9] Paster BJ, Boches SK, Galvin JL, Ericson RE, Lau CN, Levanos VA, Sahasrabudhe
A, Dewhirst FE. Bacterial diversity in human subgingival plaque. J Bacteriol
2001;183:377083.
[10] Kazor CE, Mitchell PM, Lee AM, Stokes LN, Loesche WJ, Dewhirst FE, Paster BJ.
Diversity of bacterial populations on the tongue dorsa of patients with
halitosis and healthy patients. J Clin Microbiol 2003;41:55863.
[11] Wade WG, Spratt DA, Dymock D, Weightman AJ. Molecular detection of novel
anaerobic species in dentoalveolar abscesses. Clin Infect Dis 1997;25(Suppl 2):
S2356.
[12] Kumar PS, Griffen AL, Moeschberger ML, Leys EJ. Identication of candidate
periodontal pathogens and benecial species by quantitative 16S clonal
analysis. J Clin Microbiol 2005;43:394455.
[13] Kumar PS, Leys EJ, Bryk JM, Martinez FJ, Moeschberger ML, Griffen AL.
Changes in periodontal health status are associated with bacterial commu-
nity shifts as assessed by quantitative 16S cloning and sequencing. J Clin
Microbiol 2006;44:366573.
[14] Becker MR, Paster BJ, Leys EJ, Moeschberger ML, Kenyon SG, Galvin JL, Boches
SK, Dewhirst FE, Griffen AL. Molecular analysis of bacterial species associated
with childhood caries. J Clin Microbiol 2002;40:10019.
Fig. 3. Relative proportions of 6 dominant bacterial genera in the salivary [15] Chhour KL, Nadkarni MA, Byun R, Martin FE, Jacques NA, Hunter N. Molecular
microbiota of representative samples of each cluster, as determined by clone analysis of microbial diversity in advanced caries. J Clin Microbiol
library analysis. The number of clones whose nucleotide sequences were deter- 2005;43:8439.
mined was 159, 175, and 171 in clusters I, II, and III, respectively. [16] Gross EL, Leys EJ, Gasparovich SR, Firestone ND, Schwartzbaum JA, Janies DA,
Asnani K, Griffen AL. Bacterial 16S sequence analysis of severe caries in
young permanent teeth. J Clin Microbiol 2010;48:41218.
potential pathogen, and the characteristic bacterial environment [17] Mager DL, Ximenez-Fyvie LA, Haffajee AD, Socransky SS. Distribution of
may affect gingival health through such interactions. Although a selected bacterial species on intraoral surfaces. J Clin Periodontol 2003;30:
methodology for altering oral microbiota remains to be developed, 64454.
[18] Pace NR. A molecular view of microbial diversity and the biosphere. Science
manipulation of the dominant oral microbiota (i.e., suppression or
1997;276:73440.
promotion of specic bacterial populations) represents another [19] Sakamoto M, Siqueira Jr JF, Rocas IN, Benno Y. Molecular analysis of the root
approach to oral health maintenance. canal microbiota associated with endodontic treatment failures. Oral Micro-
biol Immunol 2008;23:27581.
[20] Chen T, Yu WH, Izard J, Baranova OV, Lakshmanan A, Dewhirst FE. The human
oral microbiome database: a web accessible resource for investigating oral
5. Conclusions
microbe taxonomic and genomic information. Database (Oxford) 2010
baq013.
In this review, we describe the relationship between the oral [21] Liu WT, Marsh TL, Cheng H, Forney LJ. Characterization of microbial diversity
microbiota as a whole, obtained using a molecular ecological by determining terminal restriction fragment length polymorphisms of genes
encoding 16S rRNA. Appl Environ Microbiol 1997;63:451622.
approach, and oral health. Next-generation sequencing generates [22] Muyzer G, de Waal EC, Uitterlinden AG. Proling of complex microbial
vast amounts of sequence data at a much lower cost than populations by denaturing gradient gel electrophoresis analysis of polymer-
conventional techniques, and detailed phylogenetic information ase chain reaction-amplied genes coding for 16S rRNA. Appl Environ Microbiol
on a large number of bacterial community samples is accumulat- 1993;59:695700.
[23] Sakamoto M, Huang Y, Ohnishi M, Umeda M, Ishikawa I, Benno Y. Changes in
ing. Using this novel approach, the relationship between the oral microbial proles after periodontal treatment as determined by mole-
structure of the salivary microbiota and dental caries was cular analysis of 16S rRNA genes. J Med Microbiol 2004;53:56371.
reported [29]. A broad ecological view of our relationship with [24] Takeshita T, Nakano Y, Yamashita Y. Improved accuracy in terminal restric-
tion fragment length polymorphism phylogenetic analysis using a novel
oral microbes will provide novel insight into the most effective
internal size standard denition. Oral Microbiol Immunol 2007;22:41928.
methods of maintaining oral and, subsequently, systemic health. [25] Nakano Y, Takeshita T, Yamashita Y. TRFMA: a web-based tool for terminal
restriction fragment length polymorphism analysis based on molecular
weight. Bioinformatics 2006;22:17889.
Conict of interest [26] Takeshita T, Nakano Y, Kumagai T, Yasui M, Kamio N, Shibata Y, Shiota S,
Yamashita Y. The ecological proportion of indigenous bacterial populations in
saliva is correlated with oral health status. ISME J 2009;3:6578.
No potential conicts of interest are disclosed. [27] Kononen E, Paju S, Pussinen PJ, Hyvonen M, Di Tella P, Suominen-Taipale L,
Knuuttila M. Population-based study of salivary carriage of periodontal
pathogens in adults. J Clin Microbiol 2007;45:244651.
References
[28] von Troil-Linden B, Alaluusua S, Wolf J, Jousimies-Somer H, Torppa J,
Asikainen S. Periodontitis patient and the spouse: periodontal bacteria before
[1] Bratthall D. Demonstration of ve serological groups of streptococcal strains and after treatment. J Clin Periodontol 1997;24:8939.
resembling Streptococcus mutans. Odontol Revy 1970;21:14352. [29] Yang F, Zeng X, Ning K, Liu KL, Lo CC, Wang W, Chen J, Wang D, Huang R,
[2] Ota F, Ota M, Mahmud ZH, Mohammad A, Yamato M, Kassu A, Kato Y, Chang X, Chain PS, Xie G, Ling J, Xu J. Saliva microbiomes distinguish caries-
Tomotake H, Batoni G, Campa M. Serological diversity demonstrable by a set active from healthy human populations. ISME J 2012;6:110.

You might also like