Você está na página 1de 12

Periodontology 2000, Vol. 70, 2016, 26–37 © 2015 John Wiley & Sons A/S.

y & Sons A/S. Published by John Wiley & Sons Ltd


Printed in Singapore. All rights reserved PERIODONTOLOGY 2000

Personalized medicine: an
update of salivary biomarkers for
periodontal diseases
D I N A L. K O R T E & J A N E T K I N N E Y

For oral health-care clinicians, there can be a level of significance, as well as of panels of combined
uncertainty in predicting successful periodontal markers and periodontal pathogens that reveal
site-specific treatment outcomes and stability. When increased sensitivity and specificity for obtaining
using the conventional approach of probing depths accurate diagnostic information.
as a measure of disease progression, more than
2 mm of clinical attachment loss must occur before
a site is deemed as having ‘progressed’, making this Periodontal disease diagnosis and
method of monitoring disease breakdown an assess- monitoring
ment of disease ‘history’ rather than a ‘real time’
appraisal of disease activity (6, 7). Thus, the need Fossil evidence of alveolar bone loss surrounding the
exists for an innovative diagnostic instrument with root surfaces of teeth demonstrates that periodontal
the capability of detecting real-time changes in the disease is an ancient disease and yet remains as the
periodontium. Oral fluids, such as gingival crevicular most common cause of tooth loss in the world today
fluid and saliva, have emerged as potential supple- (19). Periodontal diseases are complex, multifactorial
mental diagnostic tools. Early investigations focusing chronic inflammatory infections which affect the
on gingival crevicular fluid sought to identify the bone and soft tissue that support the teeth (1, 5, 65).
presence and functions of proteins, particularly the Microorganisms and microbial products in the pla-
enzymes released from damaged periodontal tissue, que biofilm are the main etiologic factors that lead to
and the complex role of neutrophil-mediated host degradation of the supportive periodontal structures.
defense (21). Later investigations began exploring In addition, the host immune response, as well as
saliva as a host-derived oral fluid with potential diag- local and systemic factors, play a significant role in
nostic capabilities. Saliva is a fluid rich in serum the process of destruction or maintenance of the peri-
albumin and in antimicrobial and immunomodulary odontium. (19, 56).
proteins, which contribute to mucosal lubrication, Periodontal diseases are widely prevalent in the
tooth-structure buffering and overall maintenance of USA, where over one-half of adults are affected by
the integrity of the oral cavity (66). Studies have some level of the disease (2). A more recent preva-
unveiled the potential to identify and measure pan- lence investigation of adult periodontitis in the USA,
els of biomarkers in saliva for diagnosing periodontal with data from the 2009 and 2010 National Health
diseases and monitoring progression and health. and Nutrition Examination Survey, revealed that over
Periodontal tissue physiology and periodontal 47% of the sample had periodontitis, which equates
diseases are very complex, and have provided to 64.7 million adults (23).
challenges along the way toward reaching the goal of Many studies have been published in the past
chair-side salivary diagnostics for individualized decade that contribute to the notion that periodontal
treatment and maintaining periodontal health. This diseases may truly influence a number of systemic
review serves to provide an overview of salivary diseases, such as diabetes, cardiovascular events and
biomarkers that demonstrate potential for diagnostic osteoporosis. One hypothesis is that bacteria from

26
Salivary diagnostics

plaque biofilm enter the bloodstream and cause by factors such as time of day, duration of collection
infection and inflammation at a distant site. Another time, temperature, hydration status of the individual,
relates to the stimulation and release of proinflam- systemic health status of the person and emotional
matory cytokines or acute-phase proteins at a distant state of the person (13, 37, 38, 45, 62, 64, 81, 82).
site that may intensify or initiate a disease process. Despite these potential limitations, the analysis of
More research is needed to elucidate these associa- biomarkers in saliva has been shown for disease
tions, but links are being explored and may provide a detection, monitoring and compliance with
more thorough explanation of the oral–systemic links treatment recommendations, in both medicine and
(27, 67). The role of inflammation appears to be the dentistry (68, 69).
common denominator between periodontal diseases The term ‘biomarker’ refers to biologic substances
and some systemic diseases, which emphasizes that can be measured and evaluated to serve as
the importance of utilizing salivary diagnostics for indicators of biological health, pathogenic pro-
periodontal diseases and monitoring. cesses, environmental exposure and pharmacologic
Current clinical assessments used to determine responses to a therapeutic intervention (74, 76). The
periodontal disease severity, response to therapy study of salivary proteins and the complete human
and disease activity include: pocket depth; clinical salivary proteome provides an insight into the
attachment level; bleeding upon probing; gingival intricate cellular functional interactions that main-
inflammation; plaque presence or level of oral-hy- tain oral and general health. Thus, the biologic
giene care; suppuration; and radiographic bone loss. parameters (biomarkers) of health and disease can
An important caveat of periodontal disease diagnosis be better understood and allow better implementa-
is that only after the biologic onset of the disease tion of appropriate and personalized preventive
process will clinical assessments provide a diagnosis and therapeutic strategies to maintain optimal
of periodontal disease (42, 53). In addition, measure- health (74).
ment errors, such as probing angulation and force, A great deal of work has been dedicated to
can interfere with accurate measurements of attach- cataloging, centralizing the salivary proteome and
ment level and can be significant enough to distort annotating identified saliva proteins (74). This large
clinical treatment planning (31, 35). These clinical database is known as the Saliva Proteome Knowledge
measurements are helpful for assessment, but alone Base and is available to the public (http://www.skb.u-
are unable to determine current disease activity or cla.edu). This central database is crucial in identifica-
future risk of structure loss (86). tion of combinations of biomarker panels that may
provide distinct diagnostic information and thus
serves as a valuable resource for identifying devia-
Utility of salivary diagnostics in tions within proteins or peptides that align with oral
medicine and systemic disease and will aid in the development
of saliva-based diagnostic tests (22).
The vast amount of saliva is produced by the three The proteins found in salivary fluid and blood
pairs of major salivary glands (parotid, submandibu- plasma fluid have been compared (83). Approxi-
lar and sublingual) with lesser quantities of saliva mately 27% of proteins found in whole saliva are also
originating from hundreds of minor glands located in found in plasma. Of particular interest, almost 40% of
the buccal, labial and palatal tissues (80). Saliva also proteins that may be biomarkers for cancer,
contains nonsalivary elements, such as gingival cardiovascular disease and stroke can be found in
crevicular fluid, desquamated cells, nasopharyngeal whole saliva (44). This overlap of proteins in both
discharge, extraneous debris, and bacteria and bacte- plasma and saliva could allow for noninvasive, early
rial by-products (59, 87). detection and monitoring of oral and systemic dis-
The typical saliva flow rate varies between 800 and eases (Fig. 1).
1,500 ml/day, making it a readily available, plentiful Salivary diagnostics may become a reality in the
biological fluid. Another advantage of using saliva as near future with the development of analytical tools
a ‘real time’ diagnostic specimen is the fact that it can and reproducible biomarkers. For example, Zhang
be collected in a comfortable manner. Unlike a blood et al. (85) recently identified, with high sensitivity and
draw and the associated fear of the needle, or a urine specificity, salivary messenger RNA biomarkers that
sample and the intrusion of privacy, saliva can be are discriminatory for the detection of resectable pan-
collection in a noninvasive manner. Deviations in creatic cancer without the complication of chronic
salivary production and flow rates can be influenced pancreatitis. This is a breakthrough proof-of-concept

27
Korte & Kinney

Fig. 1. Biomarker overlap in saliva and blood plasma fluid. and secrete these proteins as saliva into the oral cavity. This
Salivary glands and their internal lobules are highly perme- free exchange of blood-derived protein molecules into the
able and are surrounded by capillaries. Cells within the oral fluid could allow for early detection and monitoring of
salivary glands absorb protein leaked from blood plasma oral and systemic diseases.

investigation for noninvasive detection of a systemic what stage of pathologic breakdown the patient is
cancer (85). currently experiencing. In the early inflammatory
In terms of cardiovascular biomarker research, the stage of the disease, numerous cytokines, such as
feasibility and utility of a programmable bio-nanochip prostaglandin E2, interleukin-1, interleukin-6 and
(P-BNC) to identify biomarkers of acute myocardial tumor necrosis factor-alpha, are released from a vari-
infarction in saliva, was investigated. Interestingly, ety of cells, such as junctional epithelia, connective
this nanotechnology was able to identify saliva-based tissue fibroblasts, macrophages and polymorphonu-
biomarker panels that had significant diagnostic capa- clear neutrophils (Fig. 2). As the disease progresses,
bility when used in conjunction with an electrocardio- powerful enzymes, such as matrix metalloproteinase-
gram, and far exceeded the screening capacity of an 8, matrix metalloproteinase-9 and matrix metallopro-
electrocardiogram alone (17). teinase-13, are released at the infected site, which
Saliva HIV tests have demonstrated comparable leads to the destruction of connective tissue collagen
accuracy to the traditional blood test (54). Oral HIV and alveolar bone loss. As the disease becomes more
tests can be a powerful tool for high-risk popula- severe, the levels of tumor necrosis factor, inter-
tions. Several commercial salivary tests are cur- leukin-1 and RANKL are elevated and ultimately
rently available within and outside the USA to mediate osteoclastogenesis and alveolar bone break-
screen for or diagnose many other conditions and down. Bone-specific biomarkers, such as pyridinoline
disease states, including hormone levels, alcohol and cross-linked carboxyterminal telopeptide of type I
drugs of abuse (55). collagen are dispersed into the surrounding tissue
and subsequently transported via the gingival crevic-
ular fluid into the periodontal pocket and finally into
Salivary biomarkers involved in the oral cavity, becoming a component of saliva (12,
periodontitis 84). Table 1 provides details of individual salivary
biomarkers that have been associated with various
When considering the periodontal pathogenic pro- stages of periodontal disease. Although individual
cesses, periodontitis can be generally divided into biomarkers have been studied as indicators of period-
three phases: inflammation; connective tissue degra- ontal disease progression, it is unlikely that one stan-
dation; and bone turnover. During each phase of the dalone biomarker will present with sufficiently high
disease, specific host-derived biomarkers have been sensitivity and specificity level to meet the criteria of
identified and therefore provide a general sense of a diagnostic tool. However, evidence points to panels

28
Salivary diagnostics

Fig. 2. Role of molecular mediators


in periodontal disease. Pathogenic
processes in periodontal disease are
characterized by the host immune
defense with the release of polymor-
phonuclear neutrophils (PMN) and
the response of inflammatory-phase
markers [interleukin-1beta (IL-1b)
and tumor necrosis factor-alpha
(TNF-a)], connective tissue degrada-
tion markers [matrix metallopro-
teinases (MMPs) and aspartate
aminotransferase (AST)] and bone
turnover markers [osteoprotegerin
(OPG) and carboxyterminal telopep-
tide of type I collagen]. LPS,
lipopolysaccharide; PGE2, prosta-
glandin E2. Adapted with permission
(57).

of salivary biomarkers and putative periodontal One proinflammatory cytokine that plays a
pathogens that may offer promising applications for paramount role in chronic inflammation is inter-
differential diagnosis, treatment planning and moni- leukin-1. Interleukin-1 is produced by several cell
toring, as well as for identification of patients at risk types, including monocytes, polymorphonu-
for future tissue destruction (46, 86). clear neutrophils, fibroblasts, epithelial cells,
endothelial cells and osteoblasts, and is involved
early in the inflammatory response by recruiting
Inflammatory markers
chemotactic cytokines to the infected periodontal
Periodontal destruction is orchestrated by a number site (32, 41). Interleukin-1 also stimulates enzymes
of mediators, including cytokines. Cytokines are necessary for the production of prostaglandin E2
soluble protein ‘messenger’ molecules that transmit and helps to control metalloproteinases and their
signals to other cells (3, 20, 73). Cytokines play a key inhibitors. Interleukin-1 induces osteoclastogenesis,
role in initiating and maintaining immune which ultimately results in permanent alveolar
and inflammatory responses by stimulating the pro- bone loss (9, 32, 58). The sum biological activity of
duction of secondary mediators. These mediators, in interleukin-1 is composed of two agonists – inter-
turn, evoke a cascade of events that amplify the leukin-1alpha and interleukin-1beta – and the
inflammatory response and induce the production of interleukin-1 receptor antagonist. In particular, sali-
enzymes that are responsible for the degrada- vary interleukin-1beta has been shown to be
tion of connective tissue and for osteoclastic bone elevated in periodontal diseases (24, 34, 75). Addi-
resorption. tional studies have found salivary interleukin-1beta

29
Korte & Kinney

Table 1. Biomarkers and their potentially destructive role in periodontal disease

Type of marker Salivary biomarker Function References

Inflammatory Interleukin-1beta Potent inflammatory stimulator. Strong correlation 16, 24, 34, 75
with periodontal disease progression
Interleukin-6 Regulates immune and inflammatory responses, 18, 26, 41, 43, 60, 63
and stimulates osteoclast differentiation. Increased
levels in periodontal disease
Macrophage inflammatory Synthesis and distribution of proinflammatory 4, 25
protein-1alpha cytokines. Increased salivary concentration
associated with periodontal disease
Tumor necrosis factor- Inhibits bone collagen synthesis. Induces 33, 46, 61
alpha collagenases
Connective tissue Matrix metalloproteinase-8 Degradation of interstitial collagens. Prevalent host 33, 39, 47, 57, 61
degradation proteinase in periodontal disease
Matrix metalloproteinase-9 Degradation of extracellular matrix proteins. 4, 39, 57, 72
Mediator of tissue destruction and immune
responses in periodontal disease
Tissue inhibitor of Inhibitor of matrix metalloproteinase-8 and 33, 34, 72
metalloproteinase-1 collagenolytic activity. Decreased levels in
periodontal disease
Aspartate Released during cell injury or death. Increased levels 15, 46, 50–52, 78, 79
aminotransferase during periodontal disease progression
Alkaline phosphatase Associated with the calcification process. Increased 46, 50, 51, 79
levels in periodontal disease
Alveolar bone Osteoprotegerin Competitively inhibits osteoclast differentiation and 4, 36, 39, 47, 57
turnover/ activity
resorption
Carboxyterminal Collagen degradation. Released during periodontal 4, 33, 39, 48, 57
telopeptide of type I disease progression
collagen
Microbial Aggregatibacter Putative periodontal pathogen associated with 25, 33
periodontal actinomycetemcomitans chronic and aggressive periodontitis. Carriers may
pathogens be at greater risk for periodontal disease
Porphyromonas gingivalis High prevalence in periodontal disease. Significant 34, 39, 50, 57, 71
predictor of periodontal disease progression.
Increased predictability with alkaline
aminotransferase and the inflammatory
biomarkers interleukin-1beta, interleukin-8 and
matrix metalloproteinase-8
Prevotella intermedia Significant predictor of periodontal disease 50, 71
progression
Tannerella forsythia High prevalence in chronic periodontitis. Increased 34, 39, 57, 71
predictability with the inflammatory biomarkers
interleukin-1beta, interleukin-8 and matrix
metalloproteinase-8
Treponema denticola High prevalence in periodontal disease. Significant 39, 57, 71
predictor of periodontal disease progression.
Increased predictability with inflammatory
biomarkers

to be strongly correlated with periodontal disease periodontal sites (16, 34). Salivary interleukin-1beta
progression and considered to be a good biomar- has also been associated, in a dose-dependent
ker for discriminating between active and inactive manner, with advanced periodontitis (34).

30
Salivary diagnostics

Interleukin-6 is a pleiotropic cytokine that, synergistic effect of interleukin-1 and tumor necrosis
similarly to interleukin-1, affects the activity of mul- factor, and bone resorption. Salivary levels of tumor
tiple physiologic and pathologic processes (49). necrosis factor-alpha indicate the presence of
Interleukin-6 plays a critical role in the regulation of generalized chronic periodontitis and have also
the immune and inflammatory responses (41, 49, demonstrated significant changes after nonsurgical
77). It is produced by several cell types, including periodontal therapy (61).
macrophages, neutrophils, keratinocytes, fibroblasts
and endothelial cells (49). Increased salivary levels
Connective tissue degradation
of interleukin-6 have been correlated with the pres-
ence of gingivitis and are present in individuals with Matrix metalloproteinase-8 is one of many matrix
periodontitis compared with healthy subjects (11, metalloproteinases. Subcategories of matrix metallo-
18, 26, 63). Salivary interleukin-6 has also been proteinases include collagenases, gelatinases, strome-
shown to stimulate osteoclast differentiation and lysins, matrilysins, membrane-type and other matrix
bone resorption, and is associated with tissue metalloproteinases. As a group, matrix metallopro-
destruction in periodontitis and peri-implant dis- teinases are part of a larger family of proteinases that
ease (43, 60). function in both normal physiologic states, such as
Macrophage inflammatory protein occurs in two health, tissue repair and remodeling, as well as in
forms, namely macrophage inflammatory protein- times of pathological disease destruction and
1alpha and macrophage inflammatory protein-1beta. progression. Proteinases are produced by host
Both macrophage inflammatory proteins are keratinocytes, fibroblasts, periodontal ligament cells,
produced by macrophages after stimulation with cementoblasts, osteoblasts and osteoclasts, as well as
bacterial endotoxins. As a whole, macrophage inflam- by inflammatory and immune cells, such as
matory proteins belong to a branch of cytokines neutrophils, monocytes, macrophages and plasma
called chemotactic cytokines or chemokines. As their cells. As a family, matrix metalloproteinases are
name suggests, these signaling proteins induce an responsible for remodeling and degradation of matrix
exacerbated inflammatory effect and are implicated components of the periodontium and also break
in the synthesis and distribution of other proinflam- down interstitial and basement membrane collagens.
matory cytokines, such as interleukin-1, interleukin-6 Matrix metalloproteinase-8, also referred to as
and tumor necrosis factor-alpha. Recently, Al-Sab- collagenase-2, is one of the most prevalent host
bagh et al. (4) shared the results of their cross-sec- proteinases found in diseased periodontal tissues and
tional study involving salivary concentrations of is linked to the degradation of interstitial collagens.
macrophage inflammatory protein-1alpha in subjects Elevated levels of salivary matrix metalloproteinase-8
with moderate to severe chronic periodontis have repeatedly demonstrated significant, positive
compared with sex- and age-matched healthy coun- correlations with increased bleeding upon probing,
terparts. The mean level of macrophage inflamma- clinical attachment level and pocket depth (33, 39, 47,
tory protein-1alpha was 18-fold higher in the 57). Moreover, significant reductions in salivary
periodontal subjects than in the healthy group, matrix metalloproteinase-8 levels have been found
demonstrating the strong ability of macrophage after periodontal scaling and root planing, suggesting
inflammatory protein-1alpha to distinguish between their potential ability to monitor periodontal disease
periodontal health and disease. activity (61).
Another important inflammatory and primary Matrix metalloproteinase-9, also called gelatinase
immune mediator is tumor necrosis factor-alpha. B, is a gelatinolytic enzyme capable of breaking down
Tumor necrosis factor-alpha is one of two proteins extracellular matrix proteins, including type IV
associated with the parent cytokine, tumor necrosis collagen. Recently, matrix metalloproteinase-9 has
factor; lymphotoxin-alpha, also known as tumor gained significant attention in medicine and appears
necrosis factor-beta, is the other protein. This key to be associated with cardiovascular disease, cancer,
biomarker is mainly produced by macrophages and multiple sclerosis and neuropsychiatric disorders,
plays a pivotal role in producing a wide range of such as schizophrenia and bipolar mood disorder.
systemic inflammatory responses. Specific to peri- Regarding periodontal disease, matrix metallopro-
odontal disease, tumor necrosis factor-alpha has the teinase-9 mediates many functions of the periodontal
ability to inhibit bone collagen synthesis and disease process, including tissue destruction and
induce collagenases, much like interleukin-1 (32, 46). immune responses (39, 57, 72). This biomarker has
Interestingly, in 1987, Stashenko et al. (75) reported a consistently demonstrated elevated salivary levels in

31
Korte & Kinney

clinically diseased conditions as well as reduced progression, of periodontal disease. Early studies
salivary levels in clinically stable conditions (4, 39). exploring the correlation between alkaline
Tissue inhibitor of metalloproteinase-1 is one of phosphatase and periodontal disease reported
four endogenous tissue inhibitors referred to as tissue associations between alkaline phosphatase and
inhibitor of metalloproteinases. The main function of pocket-depth measures and alkaline phosphatase and
tissue inhibitor of metalloproteinase-1 is to prevent levels of inflammation. Totan et al. (79) assessed the
the collagenolytic activities and function of matrix levels of alkaline phosphatase in saliva samples from
metalloproteinases 1, 2, 3, 8, 9 and 13. During periods clinically diagnosed periodontal patients. The findings
of periodontal quiescence, a fine balance between of this study showed that periodontal destruction,
low levels of matrix metalloproteinases and tissue assessed by measurement of pocket depth, gingival
inhibitor of metalloproteinase exists. However, during bleeding and suppuration, was associated with higher
episodes of disease progression, the delicate balance levels of alkaline phosphatase in saliva. The levels of
between these two enzymes is disrupted with levels salivary alkaline phosphatase were significantly
of matrix metalloproteinases escalating and causing increased in subjects with periodontal disease com-
extracellular matrix to be destroyed and ultimately pared with controls (46, 50, 51, 79).
pathologic periodontal lesions to form (34). For exam-
ple, an increase in matrix metalloproteinase levels
Alveolar bone turnover/resorption
and a decrease of tissue inhibitor of metallopro-
teinase levels triggers collagen degradation from both Osteoprotegerin plays an essential role in the devel-
connective tissue and alveolar bone (72). The salivary opment and maintenance of bone tissues. It is a
concentrations of tissue inhibitor of metallopro- glycoprotein that binds with RANKL and competi-
teinase-1 consistently differ between patients with tively inhibits osteoclast differentiation and activity,
periodontitis and control subjects, suggesting this resulting in the stimulation of osteoclasts (47). RANK
marker as a significant marker for periodontal disease and RANKL interactions have been shown to activate
detection and stability, especially in combination the proliferation, differentiation and survival of osteo-
with salivary matrix metalloproteinase-8 and carboxy- clasts, resulting in bone resorption. Osteoprotegerin
terminal telopeptide of type I collagen (33). is involved in preventing the bone-resorption pro-
Aspartate aminotransferase is a soluble cytoplasmic cess, as a neutralizing receptor for RANKL (14). The
enzyme that naturally occurs within the cell cyto- ratio of RANKL and osteoprotegerin plays a critical
plasm. However, during active phases of periodontal role in both physiologic bone reconstruction and
disease, cells die and aspartate aminotransferase is pathological bone loss (14, 36). In a cross-sectional
released into the surrounding tissues, making this study, the levels of salivary osteoprotegerin were
enzyme a viable marker of disease activity (52). shown to have a positive correlation in patients with
Chambers et al. (15) published one of the first studies increased bleeding upon probing and pocket depths,
showing a relationship between increased levels of but not with clinical attachment levels (47). Addition-
aspartate aminotransferase in gingival crevicular fluid ally, osteoprotegerin was not able to distinguish
and diseased periodontal tissues during an experi- patients with periodontal disease from control sub-
mental periodontitis model in Beagle dogs. Since that jects (47). However, in a more recent investigation,
time, additional studies have explored the relation- salivary osteoprotegerin was highly correlated with
ship between elevated levels of salivary aspartate periodontal disease status, and osteoprotegerin
aminotransferase and periodontal disease. Findings concentrations were significantly reduced after
from these studies link periodontal disease progres- periodontal therapy was provided (39). Salivary con-
sion, as defined by pocket depth, gingival bleeding centrations of osteoprotegerin were significantly
and suppuration, with increased levels of salivary lower within the clinically stable groups. These
aspartate aminotransferase (78, 79). When consider- finding suggest that osteoprotegerin, along with
ing the effects of periodontal treatment on aspartate connective tissue degradation markers, could
aminotransferase levels, there appears to be a parallel strengthen the predictability for periodontal disease
between the levels of aspartate aminotransferase and activity.
periodontal disease activity, with a marked decrease Carboxyterminal telopeptide of type I callagen
in aspartate aminotransferase levels after scaling (46, biomarkers are specific for alveolar bone deteriora-
50, 51). tion and offer great potential as an indicator differen-
Alkaline phosphatase is one of many host-derived tiating between patients with gingivitis and those
enzymes studied for its role in the detection, and patients undergoing active periodontal bone destruc-

32
Salivary diagnostics

tion. During active periodontal disease progression, tissue inhibitor of metalloproteinase-1 and carboxy-
the carboxyterminal telopeptide of type I callagen is terminal telopeptide of type I collagen (33). In the
released as a result of collagen degradation. In medi- smoker subjects, the accuracy was significantly greater
cine, the serum concentrations of carboxyterminal when evaluated for proportions of matrix metallopro-
telopeptide of type I collagen are being evaluated for teinase-8 with tissue inhibitor of metalloproteinase-1
their utility as biomarkers for monitoring metastatic and combinations of matrix metalloproteinase-8 with
bone levels of patients with prostate cancer. In den- carboxyterminal telopeptide of type I collagen
tistry, salivary levels of carboxyterminal telopeptide of (P < 0.001) (33). In another cross-sectional investiga-
type I collagen have been shown to be strongly corre- tion, matrix metalloproteinase-8 and interleukin-1-
lated with periodontal clinical measures (33). Addi- beta were found at significantly higher levels in
tionally, elevated levels of salivary carboxyterminal subjects with periodontitis compared with healthy
telopeptide of type I collagen have been found in sub- controls (47). Additionally, these two markers demon-
jects with periodontitis compared with control sub- strated a positive correlation with periodontal indices,
jects, but a stronger association can be made when bleeding upon probing, clinical attachment level and
combined with other markers, such as matrix metal- percentage of sites with pocket depth ≥4 mm (47).
loproteinase-8 (33, 48). Longitudinal analyses for periodontal pathogens
and host-response biomarkers have been investigated
for the ability to determine periodontal disease pro-
Robust panels of biomarkers in gression (39, 50, 57). The ‘red-complex’ pathogens
periodontitis (Porphyromonas gingivalis, Treponema denticola and
Tannerella forsythia) were able to predict periodontal
Advances in technologies have allowed development disease progression and demonstrated an association
of new methods for evaluating multiple salivary with the inflammatory biomarkers interleukin-1beta,
biomarkers. Recent cross-sectional and longitudinal interleukin-8 and matrix metalloproteinase-8. Certain
studies have identified potential panels of salivary putative periodontal pathogens, when elevated, were
biomarkers that may be used to identify sites with able to predict periodontal disease progression; these
active disease, periodontal stability and positive include the aforementioned ‘red-complex’ patho-
responses to therapy. The combinatorial markers are gens, as well as Fusobacterium nucleatum, Campy-
more robust in predicting periodontal disease pro- lobacter rectis and Prevotella intermedia. When
gression and stability (39). Recently, a study examined clustered together, these microbial markers and
six salivary protein biomarkers – matrix metallopro- inflammatory markers offer a strong relationship to
teinase-8, osteoprotegerin, macrophage inflamma- current disease status (39, 57). Additionally, alkaline
tory protein-1alpha, interleukin-1beta, interleukin-8 aminotransferase and P. gingivalis demonstrated
and tumor necrosis factor-alpha – associated with high levels of predictive ability for disease progression
chronic periodontitis (61). These biomarkers are (50). The microbial stand-outs in that study were
involved in inflammation, connective tissue degrada- P. gingivalis and P. intermedia for their ability to
tion and osteoclast-modulated alveolar bone predict periodontal disease progression. A novel diag-
turnover. This study demonstrated that three of the nostic approach was investigated for the combinato-
proteins – macrophage inflammatory protein-1a, rial ability of P. gingivalis, interleukin-1beta and
matrix metalloproteinase-8 and osteoprotegerin – are matrix metalloproteinase-8 to detect periodontitis
potentially useful in monitoring periodontal disease. (34). From data collected in their previous investiga-
Additionally, matrix metalloproteinase-8 was the tions, the researchers chose each of these markers as
stand out as the best biomarker indicative of response having the greatest potential to identify the periodon-
to therapy (61). Similarly, Kinney et al. (39) demon- tal pathogen burden, inflammatory response and
strated a panel of markers (namely matrix tissue degradation, respectively. This study proposed
metalloproteinase-8, matrix metalloproteinase-9, to define ranges (high, medium, low) of disease mark-
osteoprotegerin and interleukin-lbeta) that, in low ers that could be adopted universally for comparing
concentrations, predicted periodontal stability in study populations. They used a unique approach of
subjects monitored longitudinally. dividing risk into three categories based on cumula-
Other combinations of biomarkers that have been tions of the three markers and used multiplication of
identified together in significantly greater numbers in that result in a score. The three markers together
subjects with periodontal disease compared were able to reveal periodontitis more accurately
with healthy controls are matrix metalloproteinase-8, than each marker was individually (34).

33
Korte & Kinney

Additionally, Fine et al. (25) performed a longitudi- that will allow for accurate chair-side diagnostics
nal evaluation of salivary cytokines in children who and enhance individualized care (28). New tech-
developed localized aggressive periodontitis, and nologies are becoming available that are capable of
reported macrophage inflammatory protein-1alpha measuring salivary biomarker panels for accurate
as an early detection biomarker. In this report, the periodontal disease diagnosis and treatment recom-
marker was specific and sensitive in advance of mendations. The time is growing to a point where
radiographic evidence of bone loss (25). In a case– oral health-care providers will be able to utilize
control study, macrophage inflammatory protein-1al- high-throughput biomarker validation tools for
pha and osteoprotegerin were significantly increased rapid chair-side testing (Fig. 3). The focus has been
in subjects with periodontitis compared with healthy on the development of miniature-sized ‘chemical
subjects (4). Both of these markers demonstrated a processing units’ that process fluids and provide
positive correlation with clinical indices of percentage information that is relevant to the inflammatory,
bleeding upon probing, percentage pocket depth (≥4 connective tissue-degradation and bone-loss phases
and ≥5 mm) and percentage clinical attachment level. of periodontitis (28, 46). As the specific biomarkers
This cross-sectional study is important because the for periodontal disease and progression are deter-
level of the chemokine macrophage inflammatory mined through longitudinal analysis, it appears the
protein-1alpha was 18-fold higher in the periodontal technology is prepared to meet the scientific
disease group. The peptide, procalcitonin, is an estab- discovery. Both will come together to allow oral
lished serum marker for inflammation. It circulates at health-care providers to improve prevention and
very low levels in periodontal health, but increases treatment of periodontal diseases through personal-
dramatically in subjects with systemic infection. Sali- ized medicine.
vary and serum levels of procalcitonin have been
compared in type 2 diabetes patients with periodon-
tal disease to determine its value as a salivary marker Personalized medicine in
for periodontitis. Higher mean levels of salivary pro- periodontics
calcitonin were found for subjects with periodontitis
compared with healthy control subjects. This group Personalized medicine is a medical model that
also found that levels of procalcitonin were correlated uses genetic, genomic, environmental and clinical
with bleeding upon probing and hyperglycemic levels diagnostic testing to individualize patient care (28,
(glycated hemoglobin) (10). 40). This approach utilizes clinical assessment and
subclinical profiles to develop highly individualized
diagnosis, prognosis and treatment algorithms (28).
Point-of-care diagnostics Utilization of this model in oral health care, specif-
ically in periodontology, has the potential to
Investigators are continuously identifying and provide discriminating patient-stratification models
testing the complex periodontal disease ‘signatures’ to enhance personalized treatment algorithms.

Fig. 3. Salivary diagnostics at point-of-care. Whole saliva is report that is interpreted by the oral health-care provider,
an easily collected fluid containing microbial, genetic and to improve and personalize clinical decision making and
protein markers. Rapid point-of-care assays in a chair-side risk assessments for periodontal diseases. Reprinted with
processing unit could ultimately provide a biomarker permission (28).

34
Salivary diagnostics

Personalized medicine for periodontal diseases may 6. Armitage GC. The complete periodontal examination. Peri-
soon involve utilization of saliva to develop odontol 2000 2004: 34: 22–33.
7. Armitage GC. Comparison of the microbiological features
subclinical profiles, identifying and measuring
of chronic and aggressive periodontitis. Periodontol 2000
specific genotypes, phenotypes, putative pathogens, 2010: 53: 70–88.
inflammatory markers and collagen-degradation 8. Baelum V, Lopez R. Defining a periodontitis case: analysis
biomarkers to make informed clinical decisions of a never-treated adult population. J Clin Periodontol
about disease susceptibility, site-specific risk and 2012: 39: 10–19.
9. Barksby HE, Lea SR, Preshaw PM, Taylor JJ. The expanding
treatment interventions (29, 30, 70). When consid-
family of interleukin-1 cytokines and their role in destruc-
ering the possible utilization of saliva in a person- tive inflammatory disorders. Clin Exp Immunol 2007: 149:
alized model for periodontal disease, one can 217–225.
imagine implementation at multiple levels – 10. Bassim CW, Redman RS, DeNucci DJ, Becker KL, Nylen ES.
screening, disease detection, monitoring of treat- Salivary procalcitonin and periodontitis in diabetes. J Dent
ment outcomes and identification of refractory or Res 2008: 87: 630–634.
11. Becerik S, Ozsan N, Gu € rkan A, Oztu € Atilla G, Emingil
€ rk VO,
progressing cases. During the screening phase, the
G. Toll-like receptor 4 and membrane-bound CD14 expres-
use of saliva to identify patients at risk for future sions in gingivitis, periodontitis and CsA-induced gingival
disease activity opens the door for heightened risk- overgrowth. Arch Oral Biol 2011: 15: 797–819.
management strategies, preventive care and/or 12. Brinkmann O, Zhang L, Giannobile WV, Wong DT. Salivary
behavior change on the part of the patient to Biomarkers for periodontal disease diagnostics. Expert Opin
Med Diagn 2011: 5: 25–35.
prevent the onset of disease. At the diagnostic
13. Burlage FR, Pijpe J, Coppes RP, Hemels ME, Meertens H,
stage, identifying the presence of disease at the Canrinus A, Vissink A. Variability of flow rate when collect-
earliest possible stage may allow for less-invasive, ing stimulated human parotid saliva. Eur J Oral Sci 2005:
less-costly treatment. Saliva as a simple mechanism 113: 386–390.
for monitoring treatment outcomes, as well as 14. Cappellen D, Luong-Nguyen N-H, Bongiovanni S, Grenet
identification of refractory sites, additionally pro- O, Wanke C, Mira Susa M. Transcriptional program of
mouse osteoclast differentiation governed by the macro-
vides both the patient and the clinician with
phage colony-stimulating factor and the ligand for the
valuable information regarding the present state of receptor activator of NFjB. J Biol Chem 2002: 277: 21971–
the disease (76). 21982.
15. Chambers DA, Crawford JM, Mukherjee S, Cohen RL.
Aspartate aminotransferase increases in crevicular fluid
during experimental periodontitis in beagle dogs. J
Acknowledgments Periodontol 1984: 55: 526–530.
16. Chaudhari AU, Byakod GN, Waghmare PF, Karhadkar VM.
The authors thank multimedia designers, Ken Rieger Correlation of levels of interleukin-1b in gingival crevicular
and Stephanie O’Neil, for providing image design. We fluid to the clinical parameters of chronic periodontitis. J
acknowledge the administrative support of Karen Contemp Dent Pract 2011: 12: 52–59.
Gardner. We appreciate William Giannobile for criti- 17. Christodoulides N, Floriano PN, Sanchez X, Li L, Sc B,
Hocquard K, Patton A, Muldoon R, Miller CS, Ebersole JL,
cally reading the manuscript.
Redding S, Yeh C, Furmaga WB, Wampler DA, Bozkurt B,
Ballantyne CM, McDevitt JT. Programmable bio-nanochip
technology for the diagnosis of cardiovascular disease at
the point-of-care. Methodist Debakey Cardiovasc J 2012: 8:
References 6–12.
18. Costa PP, Trevisan GL, Macedo GO, Palioto DB, Souza SL,
1. Albandar JM. Underestimation of periodontitis in NHANES Grisi MF, Novaes AB Jr, Taba M Jr. Salivary interleukin-6,
surveys. J Periodontol 2011: 82: 337–341. matrix metalloproteinase-8, and osteoprotegerin in patients
2. Albandar JM, Brunelle JA, Kingman A. Destructive peri- with periodontitis and diabetes. J Periodontol 2010: 81:
odontal disease in adults 30 years of age and older in the 384–391.
United States, 1988–1994. J Periodontol 1999: 70: 13–29. 19. Darveau RP. Periodontitis: a polymicrobial disruption of
3. Alexander MB, Damoulis PD. The role of cytokines in the host homeostasis. Nat Rev Microbiol 2010: 8: 481–490.
pathogenesis of periodontal disease. Curr Opin Periodontol 20. Dehiya P, Kamal R, Gupta R, Bhardwaj R, Chaudhary K,
1994: 2: 39–53. Kaur S. Reactive oxygen species in periodontitis. J Indian
4. Al-Sabbagh M, Alladah A, Lin Y, Kryscio RJ, Thomas MV, Soc Periodontol 2013: 17: 411–416.
Ebersole JL, Miller CS. Bone remodeling-associated salivary 21. Delima AJ, Van Dyke TE. Origin and function of the cellular
biomarker MIP-1a distinguishes periodontal disease from components in gingival crevice fluid. Periodontol 2000
health. J Periodontal Res 2012: 47: 389–395. 2003: 31: 55–76.
5. Armitage GC. Development of a classification system for 22. Denny P, Hagen FK, Hardt M, Liao L, Yan W, Arellanno
periodontal diseases and conditions. Ann Periodontol 1999: M, Bassilian S, Bedi GS, Boontheung P, Cociorva D,
4: 1–6.

35
Korte & Kinney

Delahunty CM, Denny T, Dunsmore J, Faull KF, Gilligan varies by only about 2 degrees C. Arch Oral Biol 2005: 50:
J, Gonzalez-Begne M, Halgand F, Hall SC, Han X, Henson 919–922.
B, Hewel J, Hu S, Jeffrey S, Jiang J, Loo JA, Ogorzalek 38. Kavanagh DA, O’Mullane DM, Smeeton N. Variation of
Loo RR, Malamud D, Melvin JE, Miroshnychenko O, salivary flow rate in adolescents. Arch Oral Biol 1998: 43:
Navazesh M, Niles R, Park SK, Prakobphol A, Ramachan- 347–352.
dran P, Richert M, Robinson S, Sondej M, Souda P, Sulli- 39. Kinney JS, Morelli T, Braun T, Ramseier CA, Herr AE, Sugai
van MA, Takashima J, Than S, Wang J, Whitelegge JP, JV, Shelburne CE, Rayburn LA, Singh AK, Giannobile WV.
Witkowska HE, Wolinsky L, Xie Y, Xu T, Yu W, Ytterberg Saliva/pathogen biomarker signatures and periodontal dis-
J, Wong DT, Yates JR 3rd, Fisher SJ. The proteomes of ease progression. J Dent Res 2011: 90: 752–758.
human parotid and submandibular/sublingual gland sali- 40. Kornman KS, Duff GW. Personalized medicine: will
vas collected as the ductal secretions. J Proteome Res dentistry ride the wave or watch from the beach? J Dent Res
2008: 7: 1994–2006. 2012: 91(S8): 7–11.
23. Eke PI, Dye BA, Wei L, Thornton-Evans GO, Genco RJ. 41. Kwan Tat S, Padrines M, The oleyre S, Heymann D, Fortun
Prevalence of periodontitis in adults in the United States: Y. IL-6, RANKL, TNF-alpha/IL-1: interrelations in bone
2009 and 2010. J Dent Res 2012: 91: 914–920. resorption pathophysiology. Cytokine Growth Factor Rev
24. Engebretson SP, Grbic JT, Singer R, Lamster IB. GCF IL-1- 2004: 15: 49–60.
beta profiles in periodontal disease. J Clin Periodontol 2002: 42. Lang NP, Tonetti MS. Periodontal diagnosis in treated peri-
29: 48–53. odontitis. Why, when and how to use clinical parameters. J
25. Fine DH, Markowitz K, Furgang D, Fairlie K, Ferrandiz J, Clin Periodontol 1996: 23: 240–250.
Nasri C, McKiernan M, Donnelly R, Gunsolley J. Macro- 43. Liskmann S, Vihalemm T, Salum O, Zilmer K, Fischer K, Zil-
phage inflammatory protein-1alpha: a salivary biomarker mer M. Correlations between clinical parameters and inter-
of bone loss in a longitudinal cohort study of children at leukin-6 and interleukin-10 levels in saliva from totally
risk for aggressive periodontal disease? J Periodontol 2009: edentulous patients with peri-implant disease. Int J Oral
80: 106–113. Maxillofac Implants 2006: 21: 543–550.
26. Geivelis M, Turner DW, Pederson ED, Lamberts BL. Mea- 44. Loo JA, Yan W, Ramachandran P, Wong DT. Comparative
surements of interleukin-6 in gingival crevicular fluid from human salivary and plasma proteomes. J Dent Res 2010: 89:
adults with destructive periodontal disease. J Periodontol 1016–1023.
1993: 64: 980–983. 45. Louridis O, Demetriou N, Bazopoulou-Kyrkanides E. Envi-
27. Genco RJ, Borgnakke WS. Risk factors for periodontal dis- ronmental temperature effect on the secretion rate of “rest-
ease. Periodontol 2000 2013: 62: 59–94. ing” and stimulated human mixed saliva. J Dent Res 1970:
28. Giannobile WV. Salivary diagnostics for periodontal dis- 49: 1136–1140.
eases. J Am Dent Assoc 2012: 143(10 suppl): 6–11. 46. Miller CS, Foley JD, Bailey AL, Campell CL, Humphries RL,
29. Giannobile WV, Braun TM, Caplis AK, Doucette-Stamm L, Christodoulides N, Floriano PN, Simmons G, Bhagwandin
Duff GW, Kornman KS. Patient stratification for preventive B, Jacobson JW, Redding SW, Ebersole JL, McDevitt JT.
care in dentistry. J Dent Res 2013: 92: 694–701. Current developments in salivary diagnostics. Biomark Med
30. Giannobile WV, Kornman KS, Williams RC. Personalized 2010: 4: 171–189.
medicine enters dentistry: what might this mean for clinical 47. Miller CS, King CP, Langub MC, Kryscio RJ, Thomas MV.
practice? J Am Dent Assoc 2013: 144: 874–876. Salivary biomarkers of existing periodontal disease: a cross-
31. Goodson JM. Diagnosis of periodontitis by physical mea- sectional study. J Am Dent Assoc 2006: 137: 322–329.
surement: interpretation from episodic disease hypothesis. 48. Ng PYB, Donley M, Hausmann E, Hutson AD, Rossomando
J Periodontol 1992: 63: 373–382. EF, Scannapieco F. Candidate salivary biomarkers associ-
32. Graves DT, Cochran D. The contribution of interleukin-1 ated with alveolar bone loss: cross-sectional and in vitro
and tumor necrosis factor to periodontal tissue destruction. studies. FEMS Immunol Med Microbiol 2007: 49: 252–260.
J Periodontol 2003: 74: 391–401. 49. Nibali L, Fedele S, D’Aiuto F, Donos N. Interleukin-6 in oral
33. Gursoy UK, Ko € no€ nen E, Pradhan-Palikhe P, Tervahartiala T, diseases: a review. Oral Dis 2012: 18: 236–243.
Pussinen PJ, Suominen-Taipale L, Sorsa T. Salivary MMP-8, 50. Nomura Y, Shimada Y, Hanada N, Numabe Y, Kamoi K, Sato
TIMP-1, and ICTP as markers of advanced periodontitis. J T, Gomi K, Arai T, Inagaki K, Fukuda M, Noguchi T, Yoshie
Clin Periodontol 2010: 37: 487–493. H. Salivary biomarkers for predicting the progression of
34. Gursoy UK, Ko € no€nen E, Pussinen PJ, Tervahartiala T, chronic periodontitis. Arch Oral Biol 2012: 57: 413–420.
Hyva €rinen K, Suominen AL, Uitto V, Paju S, Sorsa T. Use of 51. Nomura Y, Tamaki Y, Tanaka T, Arakawa H, Tsurumoto A,
host- and bacteria-derived salivary markers in detection of Kirimura K, Sato T, Hanada N, Kamoi K. Screening of peri-
periodontitis: a cumulative approach. Dis Markers 2011: 30: odontitis with salivary enzyme tests. J Oral Sci 2006: 48:
299–305. 177–183.
35. Imrey PB, Chilton NW. Design and analytic concepts for 52. Page RC. Host response tests for diagnosing periodontal
periodontal clinical trials. J Periodontol 1992: 63: diseases. J Periodontol 1992: 63(4 Suppl): 356–366.
1124–1140. 53. Page RC, Kornman KS. The pathogenesis of human peri-
36. Jin Q, Cirelli JA, Park CH, Sugai JV, Taba M Jr, Kostenuik PJ, odontitis: an introduction. Periodontol 2000 1997: 14: 9–11.
Giannobile WV. RANKL inhibition through osteoprotegerin 54. Pant Pai N, Balram B, Shivkumar S, Martinez-Cajas JL,
blocks bone loss in experimental periodontitis. J Periodon- Claessens C, Lambert G, Peeling RW, Joseph L. Head-to-
tol 2007: 78: 1300–1308. head comparison of accuracy of a rapid point-of-care HIV
37. Kariyawasam AP, Dawes C. A circannual rhythm in unstim- test with oral versus whole-blood specimens: a systematic
ulated salivary flow rate when the ambient temperature review and meta-analysis. Lancet 2012: 12: 373–380.

36
Salivary diagnostics

55. Pfaffe T, Cooper-White J, Beyerlein P, Kostner K, Pun- ment of periodontal inflammation. Ann Med 2006: 38: 306–
yadeera C. Diagnostic potential of saliva: current state and 321.
future applications. Clin Chem 2011: 57: 675–687. 73. Souza PP, Lerner UH. The role of cytokines in inflammatory
56. Pihlstrom BL. Periodontal risk assessment, diagnosis and bone loss. Immunol Invest 2013: 42: 555–622.
treatment planing. Periodontol 2000 2001: 25: 37–58. 74. Spielmann N, Wong DT. Saliva: diagnostics and therapeutic
57. Ramseier CA, Kinney JS, Herr AE, Braun T, Sugai JV, Shel- perspectives. Oral Dis 2011: 17: 345–354.
burne CA, Rayburn LA, Tran HM, Singh AK, Giannobile WV. 75. Stashenko P, Dewhirst FE, Peros WJ, Kent RL, Ago JM. Syn-
Identification of pathogen and host-response markers cor- ergistic interactions between interleukin 1, tumor necrosis
related with periodontal disease. J Periodontol 2009: 80: factor, and lymphotoxin in bone resorption. J Immunol
436–446. 1987: 138: 1464–1468.
58. Salvi GE, Lang NP. Host response modulation in the man- 76. Taba M, Kinney J, Kim AS, Giannobile WV. Diagnostic
agement of periodontal diseases. J Clin Periodontol 2005: 32 biomarkers for oral and periodontal diseases. Dent Clin
(Suppl 6): 108–129. North Am 2005: 49: 551–571.
59. Schenkels LC, Veerman EC, Nieuw Amerongen AV. Bio- 77. Taga T, Kishimoto T. Gp130 and the interleukin-6 family of
chemical composition of human saliva in relation to other cytokines. Annu Rev Immunol 1997: 15: 797–819.
mucosal fluids. Crit Rev Oral Biol Med 1995: 6: 161–175. 78. Todorovic T, Dozic I, Vicente-Barrero M, Ljuskovic B, Pe-
60. Schierano G, Bellone G, Cassarino E, Pagano M, Preti G, jovic J, Marjanovic M, Knezevic M. Salivary enzymes and
Emanuelli G. Transforming growth factor-beta and inter- periodontal disease. Med Oral Patol Oral Cir Bucal 2006:
leukin 10 in oral implant sites in humans. J Dent Res 2003: 11: E115–E119.
82: 428–432. 79. Totan A, Greabu M, Totan C, Spinu T. Salivary aspartate
61. Sexton WM, Lin Y, Kryscio RJ, Dawson DR, Ebersole JL, aminotransferase, alanine aminotransferase and alkaline
Miller CS. Salivary biomarkers of periodontal disease in phosphatase: possible markers in periodontal diseases?
response to treatment. J Clin Periodontol 2011: 38: 434–441. Clin Chem Lab Med 2006: 44: 662–665.
62. Shannon IL, Chauncey HH. A parotid fluid collection device 80. Veerman EC, Van den Keybus PA, Vissink A, Nieuw
with improved stability characteristics. J Oral Ther Pharma- Amerongen AV. Human glandular salivas: their separate
col 1967: 4: 93–97. collection and analysis. Eur J Oral Sci 1996: 104: 346–352.
63. Shimada Y, Komatsu Y, Ikezawa-Suzuki I, Tai H, Sugita N, 81. Vissink A, Spijkervet FKL, Van Nieuw Amerongen A. Aging
Yoshie H. The effect of periodontal treatment on serum lep- and saliva: a review of the literature. Spec Care Dentist 1996:
tin, interleukin-6, and C-reactive protein. J Periodontol 16: 95–103.
2010: 81: 1118–1123. 82. Wu AJ, Baum BJ, Ship JA. Extended stimulated parotid and
64. Ship JA. Improving oral health in older people. J Am Geriatr submandibular secretion in a healthy young and old popu-
Soc 2002: 50: 1454–1455. lation. J Gerontol 1995: 50A: M45–M48.
65. Ship JA, Beck JD. Ten-year longitudinal study of periodontal 83. Yan W, Apweiler R, Balgley BM, Boontheung P, Bundy JL,
attachment loss in healthy adults. Oral Surg Oral Med Oral Cargile BJ, Cole S, Fang X, Golzalez-Begne M, Griffin TJ, Ha-
Pathol Oral Radiol Endod 1996: 81: 281–290. gen F, Hu S, Wolinsky LE, Lee CS, Malumud D, Melvin JE,
66. Ship JA, Pillemer SR, Baum BJ. Xerostomia and the geriatric Menon R, Mueller M, Qiao R, Rhodus NL, Sevinsky JR,
patient. J Am Geriatr Soc 2002: 50: 535–543. States D, Stephenson JL, Than S, Yates JR, Yu W, Xie H, Xie
67. Sima C, Glogauer M. Diabetes mellitus and periodontal dis- Y, Omenn GS, Loo JA, Wong DT. Systematic comparison of
eases. Curr Diab Rep 2013: 13: 445–452. the human saliva and plasma proteomes. Proteomics Clin
68. Slaughter YA, Malamud D. Oral diagnostics for the geriatric Appl 2009: 3: 116–134.
populations: current status and future prospects. Dent Clin 84. Yucel-Lindberg T, B age T. Inflammatory mediators in the
North Am 2005: 49: 445–461. pathogenesis of periodontitis. Expert Rev Mol Med 2013: 15:
69. Slavkin HC. Toward molecularly based diagnostics for the e7.
oral cavity. J Am Dent Assoc 1998: 129: 1138–1143. 85. Zhang L, Farrell JJ, Zhou H, Elashoff D, Akin D, Park N et al.
70. Slavkin HC. Revising the scope of practice for oral health Salivary Transcriptomic biomarkers for detection of resect-
professionals: enter genomics. J Am Dent Assoc 2014: 145: able pancreatic cancer. Gastroenterology 2010: 138:
228–230. 949–957.
71. Socransky SS, Haffajee AD. The bacterial etiology of 86. Zhang L, Henson BS, Camargo PM, Wong DT. The clinical
destructive periodontal disease: current concepts. J Peri- value of salivary biomarkers for periodontal disease.
odontol 1992: 63(4 Suppl): 322–331. Periodontol 2000 2009: 51: 25–37.
72. Sorsa T, Tjaderhane L, Konttinen YT, Lauhio A, Salo T, Lee 87. Zimmermann BG, Park NJ, Wong DT. Genomic targets in
HM, Golub LM, Brown DL, Mantyla P. Matrix metallopro- saliva. Ann N Y Acad Sci 2007: 1098: 184–191.
teinases: contribution to pathogenesis, diagnosis and treat-

37

Você também pode gostar