Sie sind auf Seite 1von 12

Oral Diseases (2011) doi:10.1111/j.1601-0825.2011.01851.

x
 2011 John Wiley & Sons A/S
All rights reserved
www.wiley.com

INVITED MEDICAL REVIEW

The oral microbiome in health and disease and the


potential impact on personalized dental medicine
MF Zarco, TJ Vess*, GS Ginsburg
Institute for Genome Sciences & Policy, Duke University, Durham, NC, USA

Every human body contains a personalized microbiome aran, 2010). The number of microbial cells within a
that is essential to maintaining health but capable of human body exceeds the total number of human cells in
eliciting disease. The oral microbiome is particularly the body by nearly 10 times (Turnbaugh et al, 2007;
imperative to health because it can cause both oral and Ling et al, 2010). These microorganisms contribute their
systemic disease. The oral microbiome rests within bio- genome, known as the metagenome, to the human body,
films throughout the oral cavity, forming an ecosystem multiplying human genes by approximately 100 times
that maintains health when in equilibrium. However, (Turnbaugh et al, 2007; Ling et al, 2010; Rajendhran
certain ecological shifts in the microbiome allow patho- and Gunasekaran, 2010). The activity of the micro-
gens to manifest and cause disease. Severe forms of oral biome and, specically, the expression of its metage-
disease may result in systemic disease at different body nome provide the human with resources and traits that
sites. Microbiomics and metagenomics are two fields of did not originally evolve with the body (Rajendhran and
research that have emerged to identify the presence of Gunasekaran, 2010). For example, the microbiome
specific microbes in the body and understand the nature contains genes that allow humans to digest certain plant
of the microbiome activity during both health and dis- polysaccharides (Rajendhran and Gunasekaran, 2010).
ease. The analysis of the microbiome and its genomes will There are various microhabitats throughout the body
pave the way for more effective therapeutic and diag- that contribute to the overall microbiome. The mouth,
nostic techniques and, ultimately, contribute to the skin, gut, etc. each contains its exclusive microbiome and
development of personalized medicine and personalized metagenome (Badger et al, 2011; Sonnenburg and Fisch-
dental medicine. bach, 2011). Each microhabitat maintains a unique
Oral Diseases (2011) doi: 10.1111/j.1601-0825.2011.01851.x ecosystem with distinct atmospheric and nutritional
compositions that provide a setting for symbiotic inter-
Keywords: microbiome; genomics; personalized dental medicine; actions among the various microbes within that ecosys-
diagnostics; metagenomics; medicine tem and the host. Of note, microbiomes from the same
location on the body are more similar among dierent
individuals than microbiomes from dierent locations on
the same individual (Sonnenburg and Fischbach, 2011).
Introduction The human microbiome can be classied into a core
Every human body contains a personalized set of microbiome and a variable microbiome (Turnbaugh
foreign inhabitants essential to maintaining health, yet et al, 2007). The core microbiome is shared among all
also capable of eliciting disease. The totality of these individuals and is comprised of the predominant species
microorganisms, their genomes and ecosystems encom- that exist under healthy conditions at dierent sites of
passes the microbiome1 (Parahitiyawa et al, 2010). The the body (Turnbaugh et al, 2007; Zaura et al, 2009;
human and its microbiome together make up a supra- Sonnenburg and Fischbach, 2011). The variable
organism (Ling et al, 2010; Rajendhran and Gunasek- microbiome is exclusive to the individual and has
evolved in response to unique lifestyle, and phenotypic
Correspondence: Prof. Georey S. Ginsburg, Center for Genomic and genotypic determinants (Figure 1). Although indi-
Medicine, Institute for Genome Sciences & Policy, Duke University viduals share microbiota at similar sites of the body,
School of Medicine, 101 Science Drive, DUMC Box 3382, Durham, there are varying dierences at the species and strain
NC 27708, USA. Tel: 919668 6210, Fax: 919668 6202, E-mail: level of the microbiome that can be as inimitable to the
georey.ginsburg@duke.edu
Received 30 July 2011; accepted 1 August 2011 individual as is the ngerprint (Dethlefsen et al, 2007).
*Current address: Ofce of Undergraduate Research, Virginia Tech, As the correlation between the human microbiome and
Blacksburg, VA, USA. health becomes more evident, microbiome research is
1
The terms microbiome, microora, and microbiota can be used becoming central to the advancement of disease diagnos-
interchangeably.
The oral microbiome
MF Zarco et al

2
The emerging eld of research that targets the microb-
iome for therapeutic purposes is known as microbiomics.
Microbiomics aims to understand how microorganisms
interplay with its hosts physiology and health by analyz-
ing their distinct functions and interrelationships (Raj-
endhran and Gunasekaran, 2010). Bacteria as a
determining factor of disease was a concept that emerged
in 1882 when physicist Robert Koch wrote an article
discussing bacteria as the etiological agents of tuberculo-
sis (Socransky and Haajee, 1992). Kochs article trig-
gered years of microbiology and disease etiology research
that eventually led to microbiomics. Figure 2 summarizes
several groundbreaking discoveries that were steps to the
study of the microbiome today (Socransky and Haajee,
1992). Today, the Human Microbiome Project (HMP)
takes a leading role in human microbiomics. It explores
the role of the human microbiome in physiology, health,
and disease through metagenomic research, which ana-
lyzes the genomes of specic microorganisms (Rajendh-
ran and Gunasekaran, 2009).
Specically, studies have shown the oral cavitys
microbiome to be a key source in the etiology of many
Figure 1 Overall human microbiome consists of a core microbiome, oral and systemic diseases (Scannapieco, 1998; Garcia
common to all individuals, and a variable microbiome, unique to et al, 2001). Because the oral microbiome is vital to a
individuals depending on lifestyle and physiological differences. bodys overall health, it has become an essential focus of
Reprinted by permission from Macmillan Publishers Ltd: Nature
(Turnbaugh et al, 2007) microbiomics. It is crucial to unravel the complexities of
the oral microbiome to learn the mechanisms by which it
maintains health or causes disease.
tics and therapeutics as well as the development of
personalized medicine (Sonnenburg and Fischbach, Microbiomics and metagenomics
2011). Because each individual harbors a unique microb-
iome that plays a key role in the etiology of disease within To truly understand the activity of the microbiome for
the body, disease may manifest and progress dierently therapeutic purposes, microbiomics must use metage-
among dierent individuals, making personalized medi- nomics to sequence and analyze bacterial genomes. The
cine imperative for optimal health care. HMP aims to use metagenomic techniques to sequence
However, microbiome research must develop a deeper 3000 microbial reference genomes isolated from various
understanding of the fundamentals and specics of sites around the human body. In doing so, it hopes to
microbial activity within the body during health and contribute to our understanding of how the microbiome
disease before it can contribute to personalized care. correlates with human health by identifying the dier-
First, there should be a clear picture of which microor- ences between bacterial genomes that encode for
ganisms exist in the body and how they aect the hosts metabolic functions vs disease processes (Rajendhran
physiology and health condition. Then, microbial char- and Gunasekaran, 2009).
acteristics of specic diseases should be studied to Genome sequencing has greatly enhanced the analysis
recognize microbiome patterns that distinguish one of pathogenic microorganisms, most of which are
disease from another. Finally, proper diagnostic meth- uncultivable (Horz and Conrads, 2007). Most meta-
ods and technologies should be developed to enable genomic methods are feasible because they are able
professionals to identify individual microbial proles to analyze extremely small sample sizes of bacterial
and treat specic microbes responsible for disease. genomes. The technique has led to the discovery of new

Figure 2 Summary of historical events that lead to the emergence of microbiomics

Oral Diseases
The oral microbiome
MF Zarco et al

3
genes, enzymes, and natural products, which may also identication techniques in case these laboratory proce-
be used for the development of novel chemicals and dures arrive in the clinical setting for an on-the-spot
pharmaceuticals in the future (Wong, 2010). microbial identication and personalized care.
There are several ways to sequence a metagenome or Microbiomics requires collaboration of metagenomics
segments of a metagenome, each technique serving a and clinical research to gather accurate and in depth
unique purpose. The most common techniques include information about the correlation between the microb-
16S ribosomal RNA sequencing, pyrosequencing, and iome and human health. Unfortunately, there are many
shotgun sequencing. Table 1 summarizes some methods existing factors that inhibit rapid progress of micro-
used to identify microbes, how they are performed, their biomics, beginning with complications in experimental
purpose, and their positive and negative aspects. As design. First of all, it is challenging to gather population
certain metagenomes have already been synthesized and sizes that are large enough for unbiased test results
the species identied, researchers can also use more (Badger et al, 2011). Second, there are factors in
conventional techniques to determine the presence of a sequencing that can contribute to bias, like dening a
microorganism. These techniques include culture-based operational taxonomic unit that may over or under-
identication, microscopy, enzyme analysis, and immu- estimate biodiversity in a sample. Researchers must
nological assays. Although less advanced, these proce- decide whether to sequence full-length genomes or
dures complement the sequencing processes by genome segments to determine diversity, or to undergo
providing information on the physical characteristics more expensive procedures such as pyrosequencing.
and additional behavioral and metabolic properties of Also, to fully measure the microbial diversity in the oral
microbes (Horz and Conrads, 2007; Filoche et al, 2010). cavity, there must be a more concrete denition of
It may be benecial or even necessary for the future species, which includes not only genotypic character-
dentist to be familiar with sequencing and the other istics, but phenotypic ones as well (Avila et al, 2009). In

Table 1 Methodologies for the detection of microbes and microbiomes

Technique Procedure Purpose Pros Cons

Metagenomics 16S rRNA Amplication and DNA sequences analyzed 1. Rapid 1. PCR subject to bias
sequencing cloning sequencing to identify species 2. Accurate 2. PCR sensitive to
of DNA segments 3. Detects viral DNA contamination
using PCR
Pyrosequencing Sequencing of small DNA sequences analyzed 1. Successfully determines 1. Expensive
DNA segments. to identify species and biodiversity 2. Does not produce
Pyrophosphates are also biodiversity 2. Subject to less bias full-length 16S rDNA
emitted as nucleotides because does not sequences necessary for
come together and require cloning taxonomic studies
synthesis DNA 3. Produces many
sequences
Shotgun Long DNA is DNA sequences analyzed 1. Less expensive 1. High error rate in
Sequencing randomly fragmented to identify which 2. Rapid constructing the
and sequenced. organisms are present 3. Forms long stands of continuous set of
Several rounds of and also to suggest the continuous DNA overlapping sequences
this are performed metabolic processes 4. Less need for human 2. Computationally
for multiple they are responsible intervention intensive
overlapping reads for in the microbial 5. Suggests information
for the target DNA. community on metabolic activity
Computer programs
use the overlapping
ends of dierent reads
to assemble them into
a continuous sequence
Conventional Culture Growth of bacteria on 1. Physical, behavioral, 1. Highly accurate 1. Time-consuming
microbiology analysis specic medium and chemical properties 2. Resistance testing is 2. Many periodontal
2. Metabolic requirements possible pathogens anaerobic
and or fastidious, many
species are uncultivable
Microscopy Observance of bacteria Insight about microbial Provides information 1. Oers limiting information
through microscope communities through on ecosystem 2. Requires special equipment
visual observance behavior 3. Time-consuming
Enzyme assay Laboratory technology Identies pathogen Rapid Less precise
measures enzyme by detecting presence
activities or absence of certain
enzyme activities
Immunoassay Tray inoculated with Identies presence of Rapid 1. Low sensitivity
antibody to detect microbes by antimicrobial 2. Specic antigenic
antigen activity of resistance or susceptibility molecules for marker
microbe pathogens are yet to
be identied

Oral Diseases
The oral microbiome
MF Zarco et al

4
terms of the oral microbiome, time and composition Also, the microbial ecosystem is disturbed by oral
face signicant challenges attributable to the highly hygiene practices. Even oral microbial colonies that are
dynamic characteristic of the oral cavity and its anaer- less susceptible to agitation experience changes attrib-
obic inhabitants (Badger et al, 2011). uted to diet, age, and health (Parahitiyawa et al, 2010),
Furthermore, Socransky and Haajee, (1992) presents as well as constant changes in pH, redox potential,
the complications researchers face when forming con- atmospheric conditions, salinity, and water activity from
clusions about oral pathogens. It is tricky to dene the saliva (Badger et al, 2011).
predominant etiological agent of a polymicrobial disease
caused by the opportunistic behavior of pathogenic Biolms and saliva
bacteria. Even in the case of a monomicrobial disease, Oral microorganisms adapt to changing environments
there is a potential to dene the wrong predominant within protective biolms (Avila et al, 2009). Biolms
species because a disease caused by a pathogen within a are the complex colonies of microorganisms that
niche may change the environment within that niche, predominate both hard and mucosal surfaces in the
leading other microorganism to proliferate in numbers oral cavity (Flemmig and Beikler, 2011). While the
that can skew test results. In addition, there is a microbial colonies play vital roles in maintaining oral
constantly growing number of newly identied peri- homeostasis, they are also signicant players in oral
odontal species, most probably because of the unique diseases (Flemmig and Beikler, 2011).
composition of every individual oral microbiome (Horz Dental plaque is a commonly known multispecies
and Conrads, 2007). Advanced technologies and scien- biolm that packs as layers onto tooth surfaces (Listgar-
tic methods are needed to accurately identify specic ten, 1976; Flemmig and Beikler, 2011). Dental plaque can
bacterial compositions characteristic to disease (Horz either entrap and prevent an existing oral pathogen from
and Conrads, 2007). ourishing, or provide a refuge for a pathogen to hide
However, the most complicated issue that arises in from salivary ow and the hosts immune system (Avila
microbiomics and metagenomics is the signicance of et al, 2009). Under healthy conditions, an ecological
studying microbes in isolation and outside the context of balance between microbe composition and activity keeps
their natural habitat within the human body (Keller and biolms healthy and stable (Flemmig and Beikler, 2011).
Ramos, 2008). To safely mimic and manipulate the Yet, detachment of biolms is necessary because patho-
microbiome for therapeutic purposes, tests must ensure gens can manifest within and cause disease (Filoche et al,
that microbes operate similarly in vitro and in silico as 2010) Both oral hygiene practices and salivary ow are
they do in vivo. responsible for this required detachment.
Saliva is crucial to the oral cavity because it plays a
key role in maintaining homeostasis and defending from
The oral cavity and its microbiome disease (Nieuw Amerongen and Veeman, 2002). While it
To understand the role of the oral microbiome within helps maintain a climate that allows biolms to ourish,
the oral cavity, it is important to analyze its funda- saliva also detaches layers of plaque and contains
mental characteristics and dynamics. The oral cavity numerous proteins, minerals, and antimicrobial enzymes
harbors over 700 species of bacteria that contribute to that control biolm build up and activity (Nieuw
the health and physiological status of the oral cavity. Amerongen and Veeman, 2002). Saliva also provides
Within the oral cavity, there are two types of surfaces nutrients that protect tooth enamel and antibodies
for which bacteria can colonize: the hard surfaces of that defend the oral cavity and the rest of the body
teeth and the soft tissue of the oral mucosa (Zaura from infection (Nieuw Amerongen and Veeman, 2002;
et al, 2009). The teeth, gingival sulcus, tongue, cheeks, Filoche et al, 2010).
hard and soft palates, and tonsils (Dewhirst et al, 2010) Studies have shown both salivary ow and microbiome
each provide enriching environments in which micro- composition to be unique to an individuals oral cavity,
bial communities can ourish. Dierent types of also suggesting that dental plaque composition and
microorganisms prefer distinct niches according to arrangement are specic to the individual (Filoche et al,
varying surface structures and functions (Aas et al, 2010). Varying plaque biomass, pH, and microbial
2005). Each niche provides the optimal conditions and response may result from or explain current health and
nutrients for its populating microbes (Avila et al, 2009). disease conditions, and also may explain why some
In fact, research has shown the maxilla, hard palate, individuals are more prone to oral diseases than others,
soft palate, and even the tongues lateral sides and despite oral hygiene habits (Filoche et al, 2010).
dorsal side each to have a dierent bacterial prole
(Aas et al, 2005). Oral microbiome of health
Furthermore, the oral microbiome is extremely The bacterial ora in a healthy oral cavity vs a diseased
dynamic because of the oral cavitys continuum with one is distinctly dierent, suggesting there may be a
the external environment (Parahitiyawa et al, 2010). prole for a core oral microbiome of health (Aas et al,
Thus, the oral microbiota has evolved skills to face 2005). According to various studies, identical bacterial
challenges that are not experienced by other microbiotas sequences have been discovered in the oral cavities
(Avila et al, 2009). The oral cavity has multiple essential of unrelated healthy individuals (Zaura et al, 2009;
functions that aect bacterial growth and activity: Bik et al, 2010). Bik et al (2010) performed a study
eating, communicating, and defending against infection. based on the largest set of near full-length sequences per

Oral Diseases
The oral microbiome
MF Zarco et al

5
healthy individual to date. The analysis identied 10 demonstrated such biodiversity to be crucial to health.
variables shared between 11 bacterial species. However, Analysis of plaque and saliva in healthy adults demon-
the same study also showed that signicant interindi- strated much more diversity than originally hypothe-
vidual dierences exist, supporting the concept of both a sized (Filoche et al, 2010). Oral microbiomes of children
core and variable microbiome within the oral cavity. suering from severe dental caries are much less diverse
Figure 3 shows the overlapping of specic bacterial than those of children with oral health (Kanasi et al,
genera that were found among the 10 samples. 2010). Asymptomatic lesions of infected root canals
This study and others oer a glimpse into the possible displayed a higher level of biodiversity than did the
microorganisms that predominate healthy oral cavities. symptomatic ones. The need for biodiversity in health
The major genera with the largest representation in may suggest that every species carries out a specic
healthy oral cavities include the following: Streptococ- function that is required to maintain equilibrium and
cus, Veillonella, Granulicatella, Gamella, Actinomyces, homeostasis within the oral cavity.
Corynebacterium, Rothia, Fusobacterium, Porphyromon- Furthermore, the relationship between the micro-
as, Prevotella, Capnocytophaga, Nisseria, Haemophilis, biome and its host during health is mutually benecial
Treponema, Lactobacterium, Eikenella, Leptotrichia, because the host is providing its microbial communities
Peptostreptococcus, Staphylococcus, Eubacteria, and with an environment in which they can ourish and, in
Propionibacterium (Aas et al, 2005; Jenkinson and turn, keep their host healthy. In health, microorganisms
Lamont, 2005; Wilson, 2005; Zaura et al, 2009; Bik prevent disease progression in several ways: they can
et al, 2010). Developing an in depth denition of health, prevent the adherence of pathogens onto specic
and understanding molecular dierences between health surfaces by occupying the niche preferred by a patho-
and disease, may give clinicians the ability to recognize gen, they can actively prevent a pathogen from occupy-
and diagnose diseases at an earlier and reversible stage ing a site, they can hinder a pathogens abilities to
(Zaura et al, 2009). multiply, and they can degrade a pathogens virulence
factors (Socransky and Haajee, 1992).
However, certain pathological changes within the
Oral symbiosis as the determining factor of microbial ecosystem may occur and cause a once-
health and disease benecial microorganism to initiate disease within the
The key to oral health is an ecologically balanced and oral cavity. Ecological shifts that cause pathological
diverse microbiome that practices commensalism within changes are: (1) a change in the relationships between
itself and mutualism with its host (Ruby and Goldner, the microbes and with the host; (2) an increase in
2007; Zaura et al, 2009; Filoche et al, 2010). Commensal relative abundance; and (3) acquisition of virulence
relationships among microbes allow them to ourish at factors2 (Parahitiyawa et al, 2010). In disease, microbes
no expense to their co-habitants and, in turn, maintain alter their relationship with their host from mutualistic
biodiversity within the oral cavity. Research has to parasitic and with other microbes from commensal to
opportunistic (Avila et al, 2009; Parahitiyawa et al,
2010). As the pathogenic bacteria ourish, the host
becomes infected or prone to infection (Ruby and
Goldner, 2007). Pathogens will grow with disregard of
its co-habiting bacteria, and any benecial bacteria will
not be able to inhibit the diseases manifestation (Nieuw
Amerongen and Veeman, 2002).
Because shifts in relationships, proportion, and viru-
lence properties of microbes seem to aect one another, it
is not always certain which ecological shift occurred rst.
It is also unclear what exactly triggers the initial ecological
shift and, in turn, catalyzes the entire cycle (Avila et al,
2009). The major factors that may be responsible for
initiating an ecological shift are poor oral hygiene,
compromised immune system, and genetics. Figure 4
illustrates the cycle of the ecological shifts the oral
microbiome may experience and the contributing factors
to these shifts, which eventually cause disease.
Poor oral hygiene is greatly responsible for the
accumulation of bacteria within biolms. Failure to
detach accumulating plaque will lead to overgrowth
of bacteria that may become pathogenic, reduce
biodiversity of the oral cavity, and ultimately cause
Figure 3 The inner circle presents the bacterial genera found in all 10
2
individuals; the second circle, bacteria present in 69 individuals; the Virulence factors are properties that bacteria acquire in becoming
third circle, bacteria present in 35 individuals; and the outer circle, pathogenic, such as bacterial toxins and surface proteins that are
bacteria present in 12 individuals. Adapted by permission from protective to the pathogen but destructive to other bacteria and the
Macmillan Publishers Ltd: The ISME Journal (Bik et al, 2010) host.

Oral Diseases
The oral microbiome
MF Zarco et al

6
diseases such as dental caries or periodontal disease For example, a person with Crohns disease, an auto-
(Zaura et al, 2009). Anaerobic microora in the crypts immune disease of the gastrointestinal tract, has a
of the tongue can also grow out of proportion and decreased abundance of Bacteroidetes in the intestines
develop halitosis, or consistent bad breath (Zaura et al, (Badger et al, 2011). A similar situation in the oral
2009). Proper oral hygiene practice is crucial because it cavity could result in a reduction of biodiversity and
is the only voluntary way to prevent oral disease. potentially lead to disease.
The presence of an immune system disorder can also Also, because an individuals genotype contributes to
cause an ecological shift in the microbiome. As the the makeup of its unique microbiome (Turnbaugh et al,
immune system regulates interactions between the 2007), ones genetic makeup could directly either
microbiome and the host, a compromised immune prevent the existence of certain benecial bacteria in
system usually disrupts mutual or commensal relation- the body or produce a bodily environment in which
ships (Badger et al, 2011). Although microbial relation- certain pathogenic species can reside. For example, 20%
ships during diseased states are parasitic, some of people are long-term carriers of Staphylococcus
pathogens can also facilitate the growth of other aureus. These people are more prone to staph infections,
pathogen species. In dental caries, Streptococcus mutans especially if the bacteria are not controlled. In addition,
is responsible, in part, for creating the lactic acid rich certain individuals may lack genes that encode for
environment in which Veillonella species thrive (Kanasi specic protective proteins and antibodies in saliva and,
et al, 2010). In biolm research, Veillonella species have thus, be more prone to plaque accumulation or cavities.
also been found to enhance the growth of S. mutans Once a pathogen possesses virulence factors, exists in
(Klutymans et al, 1997). Moreover, compromised im- abnormal proportions, and demonstrates parasitism, all
mune systems may inhibit the proper ow of saliva or of the following conditions which are required for
decrease the amount of nutrients present in saliva, disease have been satised: (1) The local environment is
allowing a buildup of dental plaque. For instance, one in which the species can express its virulence
Sjogrens syndrome is an autoimmune deciency that properties; (2) the pathogen is in numbers that exceed
attacks the exocrine glands and inhibits the ow of any the threshold for that host; (3) other bacterial species
saliva through the oral cavity, leading to dry mouth and can foster, or at least not inhibit, the diseases manifes-
further dental complications (Taubert et al, 2007). tation; and (4) the host is susceptible to this pathogen,
Although usually not obvious, genetic factors can be i.e., currently compromised immune system or specic
responsible for ecological shifts that lead to disease. genetic composition (Socransky and Haajee, 1992).
First, genetic factors could contribute to oral disease in Overall, it is crucial that there be an ecological balance
an indirect manner. An individual may have a specic among microorganisms to prevent pathological changes
genetic makeup that encodes for a permanent immune and disease from occurring. A healthy microbiome can
system disorder, which may then aect the microbiome. only be maintained with good oral hygiene and a well-
functioning immune system (Nieuw Amerongen and
Veeman, 2002).

The oral microbiome and the etiology of major


oral diseases
Oral diseases such as dental caries and periodontal
disease are among the most prevalent diseases world-
wide (Horz and Conrads, 2007; Selwitz et al, 2007),
aecting nearly all ages and geographic populations.
Hence, discovering the etiological factors responsible for
disease activation and progression will make way
for advanced methods of treatment and prevention.

Dental caries
Dental caries, also recognized as tooth decay and the
primary cause of oral pain and tooth loss, is a disease
that can begin as minor surface changes and persist until
there are lesions in the dentin (Selwitz et al, 2007). As
supragingival biolm matures on teeth, acid-producing
microbial colonies accumulate in dental plaque and
lower the pH of the oral cavity, creating an environment
in which they can thrive and produce more plaque
Figure 4 Cycle of ecological shifts in the oral microbiome that cause (Selwitz et al, 2007; Ling et al, 2010). These opportu-
disease. Poor oral hygiene, immunological disorders, and certain nistic pathogens cause dietary carbohydrates to ferment,
genetic compositions are major factors that contribute to the start of
this cycle. One condition may lead microbes to either grow abnormal producing acidic byproducts that destroy either
or acquire virulence factors, and in turn, activate the rest of the cycle the enamel of the crown or the root of the tooth. The
and eventually disease low-pH environment facilitates the diusion of calcium,

Oral Diseases
The oral microbiome
MF Zarco et al

7
phosphate, and carbonate out of teeth, which usually bacterial reserves in the mucous membranes that line the
protect the enamel from these pathogens. Although a oral cavity (Horz and Conrads, 2007). Today, the most
specic microbiome that signals dental caries is yet to be adequate treatment for periodontitis is simply reducing
found (Ling et al, 2010), the most common bacteria the number of pathogens present with antibiotics to
responsible for dental caries are S. mutans, Streptococ- maintain control of the disease. As antibiotics destroy
cus sobrinus, and Lactobacillus acidophilus (Streckfus an array of communities, they cannot be distributed
and Bigler, 2002; Selwitz et al, 2007). Dental caries is the loosely. The oral cavity should maintain certain Gram-
most preventable and reversible childhood disease, positive bacteria that shield pathogens from damaging
which can be avoided with proper oral hygiene, diet, hard and soft tissues (Van Essche et al, 2011).
and uoride exposure, which enables mineral resorption
back into the teeth (Streckfus and Bigler, 2002; Selwitz Oral cancer
et al, 2007). A third oral disease that deserves substantial attention is
oral cancer. Oral cancer is the sixth most prevalent
Periodontal disease cancer, aecting over 300 000 people each year around
Periodontal disease also results from subgingival plaque the world (Gill, 2011). There may be a correlation
accumulation that causes shifts in the microora from a between the structure and function of the oral microb-
healthy state to a diseased state (Horz and Conrads, iome and oral cancer (Meurman, 2010; Gill, 2011). The
2007; Filoche et al, 2010). Periodontal disease is a Gill Lab at the University of Rochester Medical Center
polymicrobial inammatory disorder of the periodon- has shown that bacterial cells of oral tumors can impact
tium (Pihlstrom et al, 2005). Gingivitis is the mildest signal pathways that initiate and advance oral cancer
form of periodontal disease (Horz and Conrads, 2007). (Gill, 2011); however, the sources of activation of
Microorganisms within biolms begin to form patho- cancerous and precancerous oral lesions have yet to be
genic characteristics that aggravate and iname the identied (Mehrotra and Yadav, 2006). Researchers
gingiva when disturbed by actions such as ossing should take full advantage of the ability to easily access
(Pihlstrom et al, 2005). Fortunately, gingivitis is easily the oral cavity and examine its microbiome and other
reversible with good oral hygiene (Horz and Conrads, precancerous characteristics for the purpose of early
2007). diagnosis, eective treatments and better chances of
Periodontitis, on the other hand, is a severe, irrevers- survival (Mehrotra and Yadav, 2006).
ible infection that attacks all soft tissue and bone that Most patients infected with oral cancer practice poor
support the periodontium and teeth structures (Horz oral hygiene (Meurman, 2010). In general, numerous
and Conrads, 2007). Like dental caries, multiple oppor- studies conducted around the world have shown poor
tunistic pathogens overgrow in dental plaque and these oral health and tooth loss to increase the risk of
abnormal proportions become pathogenic (Horz and gastric, pancreatic, and other cancers. Inammation is
Conrads, 2007; Van Essche et al, 2011). Microbes usually the rst symptom of compromised oral health
release proteolytic enzymes that break down host tissue and it gets worse as health regresses. Approximately
and may result in gingival inammation, loss of gingival 1520% of human tumors contain pathogenic agents
attachment, periodontal pocket formation, and alveolar derived from inammatory infections. Proper oral
bone and teeth destruction. The predominant pathogens hygiene will maintain control of such inammatory
involved in periodontitis are Aggregatibacter actinomy- agents that may contribute to oral cancer (Meurman,
cetemcomitans, Porphyromonas gingivalis, Prevotella 2010).
intermedia, Fusobacterium nucleatum, Tannerella for- Conversely, cancer can lead to poor oral health.
sythia, and Eikenella corredens, and Treponema denticola Carcinogens can introduce toxic agents into salivary
(Filoche et al, 2010; Dashi and Kadouri, 2011). uid that damage DNA, cause mutations, and damage
Periodontitis is extremely dicult to treat because of the integrity of oral cavity (Meurman, 2010). The oral
the nature of the disease, the complications of antimi- cavity reacts to the toxins with an inammatory
crobial therapy, and the lack of information on the response, which then produces the pathogenic agents
microbial interactions occurring during the disease. that contribute to tumor development, thus maintaining
Once pockets form in the periodontium, periodontitis a vicious carcinogenic cycle.
ocially becomes irreversible (Pihlstrom et al, 2005). Despite the limited research available about the
For one, the periodontium is unable to reattach to bone relationship between oral cancer and the oral microb-
once separated. Also, the causative pathogens deep iome, it is well known that the two most important risk
within the pockets become nearly impossible to target factors for oral cancer are tobacco and alcohol
with antimicrobial solutions (Horz and Conrads, 2007). (Johnson, 2001). Certain pathogenic strains of oral
Furthermore, periodontal pathogens develop virulent microorganisms tend to increase carcinogenic acetalde-
factors, like encapsulation, that make them resistant to hyde concentrations in saliva when metabolizing ethanol
antibiotics (Horz and Conrads, 2007; Van Essche et al, and tobacco smoke (Meurman, 2010; Yang et al, 2011).
2011). Pathogens hiding within plaque are one thousand However, not all who drink alcohol or smoke are subject
times more resistant to antimicrobials than those which to oral cancer. Those individuals are at higher risk. Also,
are more exposed (Van Essche et al, 2011). Even if each microbiome diers in the rate at which it metab-
pathogens are successfully targeted, there are high olizes the ethanol and tobacco compounds (Meurman,
chances of recolonization at treated sites because of 2010).

Oral Diseases
The oral microbiome
MF Zarco et al

8
inhibiting pathogen secretions of LPS and improving the
Relationships among oral disease, the oral
bodys glycemic control (Pihlstrom et al, 2005).
microbiome, and systemic diseases Poorly controlled diabetes increases the risk of
The oral cavity is the primary gateway to the human periodontal disease activation and severity, and the rate
body; therefore, microorganisms that inhabit that area of periodontal bone loss (Pihlstrom et al, 2005; Preshaw,
are very capable of spreading to dierent body sites 2009; Filoche et al, 2010) A hyperglycemic condition
(Dewhirst et al, 2010). Pathogens that originate in the can lead to a chronic inammatory-immune response
oral cavity can be frequently detected in blood cultures as that produces an excessive and deregulated amount of
they destroy and pass through oral mucous membranes inammatory mediators, such as cytokines and other
and periodontal pockets (Horz and Conrads, 2007). This enzymes (Pihlstrom et al, 2005). The excess inamma-
suggests a mechanism by which pathogens derived from tory mediators make their way into the periodontium,
a periodontal inammatory response make their way to casing periodontal detachment, pocket formation, and
tumors in the gut or pancreas. Pathogens may enter the even alveolar bone destruction (Preshaw, 2009). Diabe-
blood stream, alter proper immune responses, or pro- tes can also result in other oral complications such as
duce excessive and deregulated amounts of inammatory burning mouth syndrome, fungal infections, dental
mediators, and in turn, cause disease at dierent body caries, and salivary functional disorders (Kuo et al,
sites (Williams et al, 2008). Figure 5 illustrates a sum- 2008).
mary of the pathways that periodontal pathogens may
take to a systemic target organ. Persistent inammation Cardiovascular disease
and frequent bacterial attacks not only lead to bacter- Periodontal pathogens signal excessive amounts of
emia, but also organ abscesses and severe systemic antigens, endotoxins, cytokines, and C-reactive proteins
diseases, such as diabetes and cardiovascular disease that also contribute to cardiovascular complications
(Horz and Conrads, 2007; Williams et al, 2008; Meur- such as lipid deposition, smooth muscle proliferation,
man, 2010). This correlation further supports the and platelet aggregation (Kuo et al, 2008). Pathogens
importance of the oral microbiome to overall health. like P. gingivalis and Streptococcus sanguis have abilities
to induce platelet aggregation and accumulate as arterial
Diabetes plaque (Williams et al, 2008). Aggregatibacter actino-
Diabetes and periodontal disease hold a strong bidirec- mycetemcomitans in the periodontal pockets has also
tional relationship (Pihlstrom et al, 2005; Kuo et al, been discovered in atherosclerotic plaque (Bahekar et al,
2008; Williams et al, 2008). In one direction, the bacteria 2007). The organism accesses the circulatory system
involved in periodontal disease jeopardize the bodys through oral tissue and makes its way to the arteries
control of glycemic levels (Kuo et al, 2008). Porphyro- where it secretes LPS and inammatory-response medi-
monas gingivalis, a chief agent in periodontal disease, ators, resulting in atherothrombogenesis. As in diabetes,
produces a lipopolysaccharide (LPS) that is toxic to periodontal treatments may also alleviate cardiovascular
certain cytokine proteins that regulate insulin activity diseases (Tonetti et al, 2007; Kuo et al, 2008). The exact
under normal conditions. Other bacterial infections can pathway from cardiovascular disease to periodontal
also decrease the ability of skeletal muscles to uptake disease has yet to be established.
insulin-mediated glucose. This can produce whole body
insulin resistance (Kuo et al, 2008). Fortunately, peri- Disease manifestations in the oral cavity
odontal treatments can benet patients with diabetes by Analysis of the oral cavity and its microbiome may
become a means to diagnose systemic diseases that tend
to manifest in the periodontium (Pihlstrom et al, 2005).
Herpetic infections, leukemia, tuberculosis, and even
dermatological diseases are some examples of diseases
that present major gingival swelling, oral lesions, and
gingival discoloration because of cellular inltration
(Pihlstrom et al, 2005). These diseases may also invis-
ibly manifest in the oral cavity before any symptoms
become apparent in the body. For instance, respiratory
pathogens may colonize the mouths of individuals with
a high risk of pneumonia even when respiratory
symptoms are absent (Scannapieco, 1998). The oral
biolms serve as a reservoir for these pathogens and
contribute to the diseases progression. In fact, there is
also an altered oral microora in individuals with HIV,
as well as individuals who are pregnant, lactating, or
taking antibiotics. Although not all of these conditions
are diseases, this evidence suggests that homeostatic
alterations in the body manifest in the oral microbiome.
Figure 5 Periodonto pathogen pathways from the oral cavity to Detecting bacteria related to oral or systemic disease at
systemic organs. Adapted from (Scannapieco, 2004) early or asymptomatic stages may increase the chances

Oral Diseases
The oral microbiome
MF Zarco et al

9
of rapid disease reversal and alert the patient to practice aran, 2010) has the potential to naturally cure and
preventative measures. prevent disease at its early stages by incorporating
benecial bacteria that can reestablish an ecological
balance or enhance the biodiversity of a microora. For
Treatment and prevention methods
example, research showed that individuals with high
To maintain oral and systematic health, it is vital to amounts of Capnocytophaga ochracea had lower
protect the periodontium from pathogens that cause amounts of P. gingivalis and displayed no periodontal
inammatory infections. The foundation of periodontal disease progression. Individuals with low C. ochracea
therapy is anti-infective, non-surgical treatments aimed did exhibit disease progression. In probiotic treatment, a
to control biolms and the proliferation of pathogenic patient found to have elevated levels of P. gingivalis
bacteria within the oral cavity (Pihlstrom et al, 2005). could be given C. ochracea probiotics to reestablish a
First and foremost, practicing good oral hygiene is the healthy equilibrium before any periodontal disease
principal preventative measure of oral diseases. Dental generates. Prebiotics may also be used for similar
professionals may perform scaling and root planning to purposes. Prebiotics are oligosaccharides, or complex
remove plaque on tooth surfaces or infected tissue of the sugars, that aim to stimulate the growth of benecial
periodontal pockets. These procedures, combined with bacteria in the host (Badger et al, 2011). Both probiotics
persistent oral hygiene practices, can reduce tissue and prebiotics would strengthen the benecial micro-
inammation, pocket depths, and improve periodontal ora so the body can naturally ght o disease-causing
attachment (Pihlstrom et al, 2005). agents.
However, a clear denition of health, including the
Antibiotics composition and interrelationships of the healthy
When manual treatments are supplemented with local microbiome, is necessary before any probiotics can be
and systemic antibiotics, the oral cavity experiences a developed or used properly. In addition to the ambigu-
change in composition and abundance of various ity of health, the probiotic industry faces challenges
bacteria. (Pihlstrom et al, 2005). Local antibiotics kill that have prevented their market appearance (Klein
or freeze an array of species at diseased sites in the oral et al, 2010). First, the eects of probiotic organisms on
cavity, as well as heal oral lesions and halt plaque the host and the mechanism by which they exert these
accumulation (Horz and Conrads, 2007). Systemic eects are still uncertain (Sonnenburg and Fischbach,
drugs target pathogens at sites around the body in 2011). Blind delivery of probiotics is dangerous because
addition to the oral cavity but are limited to the species there is yet to be any true evidence of how they inuence
they target. They can also reduce any bleeding in the in vivo physiology and functionality. Second, the
periodontium. However, systemic drugs are convention- probiotic industry bears production parameters it must
ally used as a last resort in the treatment of periodontal overcome to be able to create safe and reliable probiotic
diseases for cost-eective purposes (Flemmig and Bei- substances (Klein et al, 2010). The production process
kler, 2011). would have to consider factors such as the technology
There can be variation in the pathogen and host used, temperature, and fermentations conditions, oxy-
response to dierent drugs. For example, growth of gen content, organic ingredients used, etc. Therapeutic
A. actinomycetemcomitans is inhibited by tetracyclines antibiotics and probiotics face limitations because of a
but unaected by clindamycin (Horz and Conrads, lack of knowledge of the oral microbiome and its
2007). Moreover, an individuals intestinal microbiota numerous, complex constituents.
aects the metabolism of drugs and toxins. As the
human microbiome is unique to the individual, oral Pathogen predators
vaccines maybe processed dierently in each body, Aside from mainstream antibiotics and emerging pro-
depending on both oral and gut microbial communities biotic approaches, an alternative treatment strategy
(Ferreira et al, 2010). involving biological antimicrobial agents may establish
Eective use of antibiotics in the future requires a groundbreaking approach to curing and preventing
genomic analysis of the patients oral microbiome to disease. Research has discovered bacterial lineages of
recognize the microbes that are present and to determine Bdellovibrio, Bacteriovorax, and Peredibacter, called
whether they will respond to specic treatments. There- Bdellovibrio-and-like organisms or BALOs, that serve
fore, the oral microbiome will likely play a central role as predators to kill anaerobic, Gram-negative bacteria
in the development and advancement of personalized (Dashi and Kadouri, 2011; Van Essche et al, 2011).
medicine. Because most periodontal pathogens are anaerobic,
Gram-negative bacteria, these BALOs may play a
Probiotics and prebiotics signicant role in the treatment of periodontal infections
While antibiotics are synthetic drugs that harm the (Horz and Conrads, 2007; Van Essche et al, 2011).
microora, probiotics are live microbes that are part of These highly motile species are abundant in aquatic
the natural microora. The utilization of antibiotics environments but cannot be isolated in large amounts
implies that disease is already in progress. Instead of (Van Essche et al, 2011). As BALOs do not prey on
ghting to cure disease, medicine today should focus on Gram-positive bacteria, and most benecial bacteria in
how to maintain health and prevent disease. Probiotic the oral cavity are Gram-positive, BALOs may even be
therapy or bacteriotherapy (Rajendhran and Gunasek- preferred over antibiotics when curing oral diseases.

Oral Diseases
The oral microbiome
MF Zarco et al

10
While antibiotics are non-specic and destroy an array be used for the analysis of microorganisms, but also in
of microbial communities, BALOs will only kill the the engineering of therapeutic agents needed to manip-
bad microbiome and sustain the good microbiome ulate the microbiome according to personal needs (Park
(Dashi and Kadouri, 2011; Van Essche et al, 2011). and Kim, 2008). Understanding changes in the oral
Furthermore, antibiotics cannot target pathogens deep microbiome at the early stages of chronic oral diseases
within layers of biolm. BALOs, on the other hand, can would allow clinicians to diagnose and treat an
inltrate and attack surface-attached bacteria (Dashi unhealthy oral cavity before the appearance of any
and Kadouri, 2011). They can also assist antibiotics by dental lesions or periodontal pockets (Zaura et al,
detaching biolm surfaces and exposing the present 2009). Additionally, the use of probiotics or other
pathogens (Dashi and Kadouri, 2011). Mixed micro- biological antimicrobial agents at early stages of disease
biota environments do not inhibit BALOs predation could naturally restore microbial equilibrium and, thus,
eciencies (Van Essche et al, 2011) and neither does minimize the need for antibiotics.
saliva nor high temperatures (Dashi and Kadouri, The diagram in Figure 6 represents the course of
2011); however, predator-prey interactions are highly chronic oral disease over time, the clinical tools that
BALO strain specic, which underscores the importance should be used to track the disease burden, and the
of identifying which pathogens are present for eective suggested use of probiotics and antibiotics to slow or
therapy (Van Essche et al, 2011). stop the disease process. For example, if specic
The utilization of BALOs seems to be a groundbreak- pathogens are recognized following screening methods,
ing approach to oral disease therapy, as these biological probiotics could be administered locally, according to
antimicrobial agents possess the benets of both anti- the amount and type of those pathogens present.
biotics and probiotics. Unfortunately, several parame- However, the role that each microorganism plays in
ters exist that prevent the use of BALOs in clinical disease progression or regression must be accurate and
settings. First, the tests that have been performed to well understood for safe and eective manipulation of
analyze BALOs behavior have only taken place in a microbiome (Zaura et al, 2009). In addition, using
experimental settings. The use of BALOs as therapeutics such clinical detection methods would require the
requires a better understanding of how these predators development of novel technologies, especially for
function in the physiological setting (Van Essche et al, performing on-the-spot tests. Personalized dental med-
2011). Second, high inoculum concentrations of BALOs icine that focuses on the oral microbiome will have
are more ecient in killing a substantial amount of extensive eects in health care, considering the oral
pathogens (Van Essche et al, 2011); yet, BALOs are not microbiomes importance to both oral and systemic
extracted from the environment in plentiful amounts. health.
Finally, BALOs are unable to function under anaerobic
conditions. This limitation means that BALOs would
Conclusion
not be able to prey on the anaerobic bacteria that reside
deep within periodontal pockets formed in periodontitis Although invisible to the naked eye, the microbiome
(Dashi and Kadouri, 2011). should not be underestimated as a key determinant of
health and disease. The oral microbial ecosystem is
particularly vital to maintaining both oral and overall
Personalized dental medicine
health in the body. Salivary ow and biolms on the
Because the microbiome is the biomarker of disease teeth and soft tissue maintain microbial equilibrium
activity, further research and advancements in microbi- within the oral cavity and protect pathogens from
omics and metagenomics are essential to understanding manifesting. Disturbing the homeostasis of the oral
the microbiology and etiology of oral diseases. Genomes cavity can stir pathogen activity and lead to oral disease.
collected through metagenomic techniques will not only Because the oral cavity is the primary gateway to the

Figure 6 Diagram of the course of chronic


disease over time (depicted by the red line)
and the clinical points of intervention that
might reduce disease burden, along with the
potential uses of probiotics (blue arrows).
Targeted antibiotic therapy (red arrow) might
reduce disease burden and activity (dashed
red line). Adapted from (Ginsburg and
Willard, 2009)

Oral Diseases
The oral microbiome
MF Zarco et al

11
body, severe cases of oral disease may result in the Flemmig TF, Beikler T (2011). Control of oral biolms.
spread of infection to other body sites, producing Periodontol 2000 55: 915.
systemic diseases such as cardiovascular disease or Garcia RI, Henshaw MM, Krall EA (2001). Relationship
exacerbating an already compromised immune system, between periodontal disease and systemic health. Periodon-
tol 2000 25: 2136.
as in diabetes. Practicing good oral hygiene and main-
Gill SR (2011). Gill lab: research overview. Department of
taining stable oral biolms is essential to keeping a body Microbiology & Immunology, University of Rochester
healthy and also preventing rapid spread of disease to Medical Center: Rochester, NY.
other individuals. Ginsburg GS, Willard HF (2009). Genomic and personalized
Microbiomics and metagenomics must collaborate to medicine: foundations and applications. Transl Res 154:
fully elucidate the nature of the microbiome during both 277287.
health and disease, which will, subsequently, pave the Horz H-P, Conrads G (2007). Diagnosis and anti-infective
way for more eective therapeutic and diagnostic therapy of periodontitis. Expert Rev Anti infect Ther 5: 11.
techniques. Ultimately, the analysis of the human Jenkinson HF, Lamont RJ (2005). Oral microbial communi-
microbiome will signicantly contribute to the develop- ties in sickness and in health. Trends Microbiol 13: 589595.
Johnson N (2001). Tobacco use and oral cancer: a global
ment of personalized medicine and personalized dental
perspective. J Dent Educ 65: 328339.
medicine. Kanasi E, Dewhirst FE, Chalmers NI (2010). Clonal analysis
of the microbiota of severe early childhood caries. Caries
Acknowledgements Res 44: 485497.
Keller M, Ramos JL (2008). Microbial goods for single cells
We want to thank Marla VacekBroadfoot for editorial and metagenomes. Curr Opin Microbiol 11: 195197.
assistance in preparation of the manuscript. This work was Klein M, Sanders ME, Duong T, Young HA (2010). Probi-
supported by the Center for Genomic Medicine in the Duke otics: from bench to market. Ann N Y Acad Sci 1212 (S1):
Institute for Genome Sciences and Policy. E1E14.
Klutymans J, Belkum AV, Verbrugh H (1997). Nasal carriage
of Staphylococcus aureus: epidemiology, underlying mecha-
Author contributions nisms, and associated risks. Clin Microbiol Rev 10: 505520.
Zarco researched the topic, wrote the initial draft, and edited Kuo L-C, Polson AM, Taeheon K (2008). Associations
the nal draft. Vess assisted Zarco in the research, and edited between periodontal diseases and systematic diseases: a
early and late drafts. Ginsburg initiated the idea, guided Zarco review of the inter-relationships and interactions with
and Vess in their research and writing, and edited and diabetes, respiratory disease, cardiovascular diseases and
approved the nal manuscript. osteoporosis. Publ Health 122: 17.
Ling Z, Kong J, Jia P et al (2010). Analysis of oral microbiota
in children with dental caries by PCR-DGGE and barcoded
References pyrosequencing. Microb Ecol 60: 677690.
Listgarten MA (1976). Structure of the microbial ora
Aas JA, Paster BJ, Stokes LN, Olsen I, Dewhirst FE (2005). associated with periodontal health and disease in man. A
Dening the normal bacterial ora of the oral cavity. J Clin light and electronmicroscopic study. J Periodontol 47: 118.
Microbiol 43: 12. Mehrotra R, Yadav S (2006). Oral squamous cell carcinoma:
Avila M, Ojcius DM, Yilmaz O (2009). The oral microbiota: etiology, pathogenesis and prognostic value of genomic
living with a permanent guest. DNA Cell Biol 28: 7. alterations. Indian J Cancer 43: 6066.
Badger JH, Ng PC, Venter JC (2011). The human genome, Meurman JH (2010). Oral microbiota and cancer. J Oral
microbiomes, and disease. In: Nelson KE, ed. Metagenomics Microbiol 2: 5195.
of the human body. Springer Science + Business Media: New Nieuw Amerongen AV, Veeman ECI (2002). Saliva the
York, vol 17, pp. 114. defender of the oral cavity. Oral Dis 8: 1222.
Bahekar AA, Singh S, Saha S, Molnar J, Arora R (2007). The Parahitiyawa NB, Scully C, Leung WK, Jin LJ, Samaranayake
prevalence and incidence of coronary heart disease is LP (2010). Exploring the oral bacterial ora: current status
signicantly increased in periodontitis: a meta-analysis. and future directions. Oral Dis 16: 10.
Am Heart J 154: 830837. Park S-Y, Kim G-J (2008). A biological teasure metagenome:
Bik EM, Long CD, Armitage GC et al (2010). Bacterial pave the way for big science. Indian J Microbiol 48: 163
diversity in the oral cavity of ten healthy individuals. ISME 172.
J 4: 962974. DOI: 10.1038/ismej.2010.30. Pihlstrom BL, Michalowicz BS, Johnson NW (2005).
Dashi A, Kadouri DE (2011). Predation of Oral Pathogens Periodontal Diseases. Lancet 366: 18091820.
by Bdellovibrio bacteriovorus 109J. Mol Oral Microbiol 26: Preshaw PM (2009). Periodontal disease and diabetes. J Dent
16. 37: S577S584.
Dethlefsen L, McFall-Ngai M, Relman DA (2007). An Rajendhran J, Gunasekaran P (2009). Human genomics an
ecological and evolutionary perspective on human-microbe microbiomics: the post-genome scenario. Curr Sci 97: 140
mutualism and disease. Nature 449: 811818. 141.
Dewhirst FE, Chen T, Izard J et al (2010). The human oral Rajendhran J, Gunasekaran P (2010). Human microbiomics.
microbiome. J Bacteriol 192: 50025017. Indian J Microbiol 50: 109112.
Ferreira RBR, Antunes LCM, Finlay BB (2010). Should the Ruby J, Goldner M (2007). Nature of symbiosis in oral
human microbiome be considered when developing vac- disease. J Dent Res 86: 811.
cines? PLoS Pathog 6: 12. Scannapieco FA (1998). Periodontal disease as a potential risk
Filoche S, Wong L, Sissons CH (2010). Oral biolms: factor for systemic diseases. J Periodontol 69: 841850.
emerging concepts in microbial ecology. J Dent Res 89: Scannapieco FA (2004). Gingivitis: an inammatory peri-
818. odontal disease. Compend Contin Educ Dent 25: 1625.

Oral Diseases
The oral microbiome
MF Zarco et al

12
Selwitz RH, Ismail AI, Pitts NB (2007). Dental caries. Lancet Van Essche M, Quirynen M, Sliepen I et al (2011). Killing of
369: 5159. anaerobic pathogens by predatory bacteria. Mol Oral
Socransky SS, Haajee AD (1992). The bacterial etiology of Microbiol 26: 5261.
destructive periodontal disease: current concepts. J Period- Williams RC, Barnett AH, Claey N et al (2008). The
ontol 63: 322331. potential impact of periodontal disease on general health:
Sonnenburg JL, Fischbach MA (2011). Community health a consensus view. Curr Med Res Opin, Infoma UK Ltd. 24:
care: therapeutic opportunities in the human microbiome. 16351643.
Sci Transl Med 3: 15. Wilson M (2005). Microbial inhabitants of humans: their
Streckfus C, Bigler L (2002). Saliva as a diagnistic uid. Oral ecology and role in health and disease. Cambridge University
Dis 8: 6976. Press: Cambridge.
Taubert M, Davies EMR, Back I (2007). Dry mouth. BMJ Wong DWS (2010). Applications of metagenomics for indus-
334: 534. trial bioproducts. In: Marco D, ed. Metagenomics: theory,
Tonetti M, DAiuto F, Nibali L et al (2007). Treatment of methods and applications. Caister Academic Press: Norfolk,
periodontitis and endothelial function. N Engl J Med 356: UK, pp. 141158.
911920. Yang L, Ganly I, Morris L et al (2011). Relevance of
Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, microbiome to cigarette smoking and oral cancer. IADR
Knight R, Gordon JI (2007). The human microbiome General Session & Exhibition, International Association for
project: a strategy to understand the microbial components Dental Research: San Diego, CA.
of the human genetic and metabolic landscape and how they Zaura E, Keijser BJ, Huse SM, Crielaard W (2009). Dening
contribute to normal physiology and predisposition to the healthy core microbiome of oral microbial commu-
disease. Nature 449: 804810. nities. BMC Microbiol 259: 12.

Oral Diseases

Das könnte Ihnen auch gefallen