Dental caries is the most prevalent and costly oral infectious disease worldwide. Virulent biofilms firmly attached to tooth surfaces are prime biological factors associated with this disease. The formation of an exopolysaccharide-rich biofilm matrix, acidification of the milieu and persistent low pH at the tooth-biofilm interface are major controlling virulence factors that modulate dental caries pathogenesis. Each one offers a selective therapeutic target for prevention. Although fluoride, delivered in various modalities, remains the mainstay for the prevention of caries, additional approaches are required to enhance its effectiveness. Available antiplaque approaches are based on the use of broad-spectrum microbicidal agents, e.g. chlorhexidine. Natural products offer a rich source of structurally diverse substances with a wide range of biological activities, which could be useful for the development of alternative or adjunctive anticaries therapies. However, it is a challenging approach owing to complex chemistry and isolation procedures to derive active compounds from natural products. Furthermore, most of the studies have been focused on the general inhibitory effects on glucan synthesis as well as on bacterial metabolism and growth, often employing methods that do not address the pathophysiological aspects of the disease (e.g. bacteria in biofilms) and the length of exposure/retention in the mouth. Thus, the true value of natural products in caries prevention and/or their exact mechanisms of action remain largely unknown. Nevertheless, natural substances potentially active against virulent properties of cariogenic organisms have been identified. This review focuses on gaps in the current knowledge and presents a model for investigating the use of natural products in anticaries chemotherapy.

The use of natural products to prevent and/or treat oral maladies dates back several thousand years both in Western and Eastern societies. For example, a large number of recipes for mouthwashes and dentifrices can be found in the Ebers Papyrus, which are composed of natural substances [Hirschfeld, 1939]. In addition, chewing stick, introduced by the Babylonians in 5000 B.C., and a wide range of traditional medicinal plants continue to be used among many African and Asian communities [Jagtap and Karkera, 2000; Pai et al., 2004; Song et al., 2007]. The term ‘natural products’ is usually associated with secondary metabolites produced by an organism, which in most cases function as defense mechanisms against herbivores, microorganisms, insects and competing plants [Singer et al., 2003]. Plants produce a diverse array of more than 100,000 secondary metabolites, and estimates of the total number in plants exceed 500,000 [Singer et al., 2003]. Secondary metabolites can be classified on the basis of composition (e.g. whether or not they contain nitrogen), the pathway by which they are synthesized, or chemical structure (e.g. the presence of rings with or without sugar moieties) [Chinou, 2008]. A simple classification includes three main groups: (i) phenolic compounds, which are made from simple sugars, containing benzene rings, hydrogen and oxygen; (ii) terpenoids, which are made from mevalonic acid and composed almost entirely of carbon and hydrogen; and (iii) alkaloids, which are nitrogen-containingcompounds [Chinou, 2008]. These chemical groups have been associated with biological effects of natural products, which may include antimicrobial, antioxidant, and anticancer activities [Newman and Cragg, 2007]. In this review, we will focus on natural products that harbor such bioactive molecules, which may exert anticaries properties.

Natural products have been used as a major source of innovative and effective therapeutic agents throughout human history, offering a diverse range of structurally distinctive bioactive molecules. Of the 1,184 new chemical entities introduced between 1981 and 2006, about half (48%) were natural products, semisynthetic natural product analogues or synthetic compounds based on natural-product pharmacophores [Newman and Cragg, 2007]. More importantly, over 70% of the therapeutic agents developed between 1981 and 2006 for infectious diseases (both bacterial and fungal) were derived from natural products [Newman and Cragg, 2007]. Naturally occurring compounds possess structures that have high chemical diversity and biochemical specificity, as well as other molecular properties that make them attractive candidates for drug discovery and development [Koehn and Carter, 2005]. Yet, studies using natural products to prevent or treat oral diseases such as dental caries have received minimal attention compared with other fields in medicine. Furthermore, many published studies are lacking critical information such as chemical characterization of the natural extracts used, and/or utilize experimental approaches that are poorly designed (see also the Caries Research policy on papers concerning anticaries effects of natural products).

Despite the widespread use of different sources of fluoride, dental caries continues to be the single most prevalent and costly oral infectious disease worldwide [National Institutes of Health, 2001; Marsh, 2003; Dye et al., 2007]. Virulent biofilms that are tightly adherent to oral surfaces are a primary cause of infectious diseases in the mouth, including dental caries [Bowen and Koo, 2011]. Dental caries results from the interactions of specific bacteria and their metabolic/virulence products with salivary constituents and dietary carbohydrates that occur on the susceptible tooth surface. The formation of the extracellular polysaccharide (EPS)-rich biofilm matrix, acidification of the milieu and the maintenance of a low-pH environment at the tooth-biofilm interface are major controlling virulence factors that modulate dental caries pathogenesis. Biofilms formed in vivo are comprised of mixed flora, although mutans streptococci are recognized as the primary producers of the EPS-rich matrix. Streptococcus mutan s plays a key role in the development of virulent biofilms, although additional microorganisms may be also involved in the pathogenesis of the disease [Beighton, 2005]. This bacterium (i) effectively utilizes dietary sucrose (and possibly starch) to rapidly synthesize EPS through the activity of glucosyltransferases (Gtfs) and a fructosyltransferase that are adsorbed to saliva-coated tooth enamel surfaces, (ii) adheres tenaciously to glucan-coated surfaces, and (iii) is both acidogenic and acid tolerant [Bowen, 2002; Quivey et al., 2000]. Thus, S. mutans thrives in the complex oral microbiome and effectively modulates the transition from nonpathogenic to cariogenic biofilms.

Cariogenic biofilms develop following initial microbial attachment to and further accumulation on the tooth surface, which is predominantly mediated by sucrose-dependent mechanisms. The EPS is mainly comprised of glucans, which are synthesized by Gtfs present in saliva, in the acquired pellicle (primarily GtfC), and those adsorbed on bacterial surfaces (mostly GtfB) in the presence of sucrose. The glucans formed in situ provide (i) avid binding sites for colonization by S. mutans and other acidogenic/aciduric organisms, and (ii) a matrix that holds the microbial cells together to form structurally cohesive cell clusters known as microcolonies [Bowen and Koo, 2011] (fig. 1). If these biofilms are not removed from the tooth surface and are frequently exposed to dietary carbohydrates (especially sucrose), S. mutans (and other acidogenic and aciduric bacteria) within the biofilm community will metabolize sucrose to organic acids and synthesize polysaccharides in situ. The elevated amounts of EPS enhance bacterial adherence and biofilm cohesiveness, shelter matrix-encased bacteria from environmental assaults (e.g. antimicrobials), improve the stability of the structure and affect the diffusion properties of the biofilm matrix [Paes Leme et al., 2006; Bowen and Koo, 2011]. Furthermore, insoluble EPS provides a framework for the establishment of tightly adherent three-dimensional biofilm structures [Koo et al., 2010b]. Conversely, soluble glucans, fructans and intracellular polysaccharides serve as short-term storage compounds that can be metabolized to increase overall acid production. Acidification of the biofilm matrix affords a competitive ecological advantage to acid stress-tolerant and acidogenic flora, such as S. mutans [Quivey et al., 2000; Marsh, 2003; Lemos and Burne, 2008]. The low-pH environment subsequently created at the tooth-biofilm interface results in demineralization of the enamel. Clearly, exopolysaccharides, acidification of the biofilm matrix as well as acid tolerance mechanisms are critical for the development of cariogenic biofilms, and thus could be attractive and specific targets for precise and highly selective chemotherapeutic strategies.

Fig. 1

Cariogenic biofilm formation (a–d) and potential targets (e) for disruption by natural products. a The Gtfs secreted by S. mutans are incorporated into pellicle (particularly GtfC) and adsorb on bacterial surfaces (mainly GtfB), including microorganisms that do not produce Gtfs (e.g. Actinomyces spp.). b Surface-adsorbed GtfB and GtfC rapidly utilize dietary sucrose (and starch hydrolysates) to synthesize insoluble and soluble glucans in situ; the soluble glucans formed by GtfD could serve as primers for GtfB enhancing overall synthesis of EPS. The Gtfs adsorbed onto enamel and microbial surfaces provide in situ an insoluble matrix for dental plaque-biofilm. Concomitantly, dietary carbohydrates (CHO) are metabolized into acids by acidogenic/aciduric organisms (e.g. S. mutans). c The glucan molecules provide avid binding sites on surfaces for S. mutans and other microorganisms mediating tight bacterial clustering and adherence to the tooth enamel (through glucan-glucan and glucan-Gbp interactions). Furthermore, Gtf-adsorbed bacteria de facto become glucan producers binding to tooth and microbial surfaces by the same mechanisms. This model could explain the rapid formation and accumulation of highly cohesive-adherent plaque in the presence of sucrose (and possibly starch) even if the number of S. mutans is relatively low. After the establishment of a glucan-rich biofilm matrix, ecological pressure (e.g. pH) will determine which bacteria may survive and dominate within plaque under frequent sucrose (or other fermentable carbohydrate) exposure. d If biofilm remains on tooth surfaces with frequent consumption of high-carbohydrate diet (especially sucrose), the amount of EPS and extent of acidification of the matrix will be increased. Such conditions cause biochemical, ecological and structural changes favoring the survival and dominance of highly acid stress-tolerant organisms in cohesive and firmly attached biofilms. The low-pH environment at the tooth-biofilm interface results in enhanced demineralization of enamel.

Fig. 1

Cariogenic biofilm formation (a–d) and potential targets (e) for disruption by natural products. a The Gtfs secreted by S. mutans are incorporated into pellicle (particularly GtfC) and adsorb on bacterial surfaces (mainly GtfB), including microorganisms that do not produce Gtfs (e.g. Actinomyces spp.). b Surface-adsorbed GtfB and GtfC rapidly utilize dietary sucrose (and starch hydrolysates) to synthesize insoluble and soluble glucans in situ; the soluble glucans formed by GtfD could serve as primers for GtfB enhancing overall synthesis of EPS. The Gtfs adsorbed onto enamel and microbial surfaces provide in situ an insoluble matrix for dental plaque-biofilm. Concomitantly, dietary carbohydrates (CHO) are metabolized into acids by acidogenic/aciduric organisms (e.g. S. mutans). c The glucan molecules provide avid binding sites on surfaces for S. mutans and other microorganisms mediating tight bacterial clustering and adherence to the tooth enamel (through glucan-glucan and glucan-Gbp interactions). Furthermore, Gtf-adsorbed bacteria de facto become glucan producers binding to tooth and microbial surfaces by the same mechanisms. This model could explain the rapid formation and accumulation of highly cohesive-adherent plaque in the presence of sucrose (and possibly starch) even if the number of S. mutans is relatively low. After the establishment of a glucan-rich biofilm matrix, ecological pressure (e.g. pH) will determine which bacteria may survive and dominate within plaque under frequent sucrose (or other fermentable carbohydrate) exposure. d If biofilm remains on tooth surfaces with frequent consumption of high-carbohydrate diet (especially sucrose), the amount of EPS and extent of acidification of the matrix will be increased. Such conditions cause biochemical, ecological and structural changes favoring the survival and dominance of highly acid stress-tolerant organisms in cohesive and firmly attached biofilms. The low-pH environment at the tooth-biofilm interface results in enhanced demineralization of enamel.

Close modal

Despite an increased understanding of the biological and physicochemical processes involved in the pathogenesis of dental caries, the vast majority of the chemical agents aiming at controlling dental biofilm formation are broad-spectrum nonspecific microbicidal agents. Chlorhexidine and triclosan are the most commonly used and clinically tested agents for antiplaque therapies [Axelsson, 1993; Eley, 1999]. There are several review articles that examine the advantages and disadvantages of these agents [Eley, 1999; Twetman, 2004], which will not be discussed in this review.

We have attempted to provide a brief summary of the current, albeit limited, state of knowledge concerning the potential biological effects of natural products on three relevant categories: (i) antimicrobial activity, specifically the inhibition of growth and metabolism of acidogenic and aciduric organisms, (ii) inhibition of exopolysaccharide synthesis, and (iii) inhibition of bacterial adherence.

Antimicrobial Activity

Inhibitors of Bacterial Growth

The majority of the studies conducted thus far have centered on investigating the effects of natural products on the growth of cariogenic microorganisms, primarily mutans streptococci (table 1). The focus on the development of bacteriostatic or bactericidal therapies is not surprising as dental caries is an infectious disease and there is an established and successful history of discovery of novel antibiotics as a result of natural product screening. Most of these compounds are derived from secondary metabolites of plant origin, including phenolic acids, anthraquinones, flavonoids, stilbenes, tannins, terpenoids and alkaloids, all of which are effective inhibitors of the growth of other pathogenic microorganisms [Wen et al., 2004]. Compounds targeting bacterial viability are typically aiming at bacterial eradication via one or more of the following modalities [Cowan, 1999]: (i) disruption of cell wall biosynthesis and/or cell membrane permeability, (ii) complexing with surface-adsorbed components, (iii) inhibiting protein synthesis or nucleic acid metabolism, and/or (iv) inhibition of enzyme activity through oxidation of other, often uncharacterized, interactions with bacterial proteins. Unfortunately, all of the modalities are generally not selective for specific species and possess broad-spectrum antibacterial effects. Table 1 lists several natural extracts and their putative active compounds which show bacteriostatic or bactericidal effects as determined using classical assays such as minimum inhibitory concentration (MIC) and minimum bacterial concentration (MBC) for batch or plate culture planktonic cells.

Table 1

Biological activities of natural products with potential use as alternative or adjunctive anticaries chemotherapy

Biological activities of natural products with potential use as alternative or adjunctive anticaries chemotherapy
Biological activities of natural products with potential use as alternative or adjunctive anticaries chemotherapy
Biological activities of natural products with potential use as alternative or adjunctive anticaries chemotherapy
Biological activities of natural products with potential use as alternative or adjunctive anticaries chemotherapy
Biological activities of natural products with potential use as alternative or adjunctive anticaries chemotherapy
Biological activities of natural products with potential use as alternative or adjunctive anticaries chemotherapy

Camellia sinensis (used to make green tea and oolong tea) has been widely studied for its antibacterial activities against caries-related bacteria, using both in vitro and in vivo models [Otake et al., 1991; Shapiro et al., 1994; Shapiro and Guggenheim, 1995; Ooshima et al., 1998]. Specific catechins from green tea have been associated with antibacterial activity against S. mutans and Streptococcus sobrinus [Sakanaka et al., 1989]. This inhibitory effect appears to be related to the presence of three hydroxy moieties at 3′, 4′ and 5′ on the B ring of the catechin and epicatechin molecular structure [Sakanaka et al., 1989]. However, the exact mechanisms of action and their putative target(s) remain to be elucidated.

Essential oils have also been extensively studied for antimicrobial activity against caries-related bacteria. Essential oils derived from plants are typically a complex mixture of approximately 20–60 compounds that are in solution at various concentrations. Overall, the main chemical group is primarily composed of terpenoids, followed by aromatic and aliphatic constituents [Bakkali et al., 2008]. Thymol and eugenol inhibit the growth of a wide range of oral microorganisms including mutans streptococci [Shapiro et al., 1994; Shapiro and Guggenheim, 1995]. In general, essential oils would appear to affect bacterial viability by compromising the integrity of the bacterial membrane. For example, thymol induces the rapid efflux of intracellular bacterial components, which likely results from the perforation of the cellular membrane [Shapiro and Guggenheim, 1995].

Propolis, a natural beehive product, and cacao bean husk extracts have also shown significant antibacterial activity against S. mutans and/or S. sobrinus in vitro [Ikeno et al., 1991; Park et al., 1998; Ooshima et al., 2000; Osawa et al., 2001]. Specific flavanones (pinocembrin), dihydroflavonols (pinobanksin-3-acetate) and terpenoids (tt- farnesol) were associated with the bioactivity of propolis [Koo et al., 2002c]. tt-Farnesol has been demonstrated to be particularly effective against both planktonic and biofilm-associated S. mutans cells [Koo et al., 2003]. Oleic and linoleic acids, a monounsaturated ω–9 fatty acid and an unsaturated ω–6 fatty acid, respectively, were identified as the main bactericidal agents in cacao extract [Osawa et al., 2001]. The antibacterial activity of tt-farnesol, oleic and linoleic acids may be related to their lipophilic characteristics and membranotropic effects, which likely damage the integrity of the cell membrane [Cowan, 1999; Osawa et al., 2001; Inoue et al., 2004] and increase proton permeability [Jeon et al., 2009].

There are many other reports in the literature concerning the antimicrobial activities that various plant extracts may have against cariogenic bacteria, although the majority of these studies provide limited or incomplete information due to the lack of chemical characterization of the extracts. However, there are a few exceptions. For example, Wu-Yuan et al. [1988] and Li et al. [1997] have identified gallotannins from Melaphis chinensis and triterpenes (ceanothic acid and ceanothetric acid) from Ceanothus americanus as antimicrobial agents that harbor activity against mutans streptococci. Furthermore, a chemically characterized extract of Galla chinensis (containing gallic acid and methyl gallate) has been demonstrated to impede the growth of S. mutans and other caries-related organisms, including Lactobacillus rhamnosus and Actinomyces naeslundii, within biofilms [Xie et al., 2008]. Recently, Greenberg et al. [2008] established that naturally occurring phenolic compounds generally display antibacterial activity by disrupting the membrane lipid-protein interface as nonionic surface-active agents.

Inhibitors of Acid Production and Acidurance

Several studies have highlighted a range of natural products that inhibit acid production in cariogenic streptococci (table 1). However, these inhibitory effects appear to be related to a reduction in bacterial growth rather than the specific inhibition of the glycolytic activity and/or aciduricity of the cariogenic organisms. Due to the limitations of the experimental procedures, compounds with specific activities were not identified (see Problems and Limitations section).

Nevertheless, recent studies have shown that some of the agents affect the acidogenicity of S. mutans by targeting multiple mechanisms, sometimes simultaneously, including (i) disruption of the membrane proton motive force, (ii) enzymatic activity and expression of specific enzymes related to sugar transport, (iii) glycolysis, and/or (iv) general metabolism. For example, 7-epiclusianone (a prenylated benzophenone isolated from Rheedia gardneriana), tt-farnesol and some cranberry flavonoids (e.g. myricetin, procyanidin A2 and A-type oligomers) have been reported to increase the proton permeability of S. mutans cells, causing cytoplasmic acidification, and thereby inhibiting the acid-sensitive intracellular glycolytic enzymes [Gregoire et al., 2007; Murata et al., 2008; Koo et al., 2010a]. Combining flavonoids (apigenin or myricetin) with tt-farnesol also disrupts the accumulation of intracellular iodophilic polysaccharides (IPS) by S. mutans in biofilms [Koo et al., 2003; Jeon et al., 2009]. IPS (a glycogen-like storage polymer) provides an endogenous source of carbohydrates for S. mutans and other cariogenic organisms, which can be metabolized when exogenous fermentable substrates have been depleted in the oral cavity, thus lowering the fasting pH in the plaque matrix. More importantly, IPS production has also been associated with dental caries development in both animals and humans [Loesche and Henry, 1967; Tanzer et al., 1976; Spatafora et al., 1995]. Recently, a crude extract of Psidium cattleianum and epigallocatechin gallate (from green tea) have been shown to disrupt the expression and activity of specific enzymes involved in the glycolytic pathway of S. mutans, including lactate dehydrogenase [Hirasawa et al., 2006; Brighenti et al., 2008]. Although these observations are promising, additional studies are needed to elucidate the mechanisms by which these agents impair the acidogenicity and aciduricity of cariogenic streptococci and lactobacilli.

Inhibition of Exopolysaccharide Synthesis

One approach to reducing the incidence of dental caries is the discovery and development of therapeutic agents that may prevent the formation of the plaque EPS-rich matrix. EPS is largely comprised of glucans synthesized by microbial Gtfs, which use dietary sucrose as their primary substrate, and Gtfs are proven virulence factors implicated in the pathogenesis of dental caries [Paes Leme et al., 2006; Bowen and Koo, 2011]. Therefore, we will focus on the activity and inhibition of the Gtfs for the purposes of this review. S. mutans, a key contributor to EPS synthesis, produces at least three distinct Gtfs – GtfB, C and D – which synthesize water-insoluble glucans rich in α-1,3-linked glucose (GtfB and C) and water-soluble glucans with mostly α-1,6-linked glucose (GtfC and D) [Loesche, 1986]. Each of the Gtfs and their polysaccharide products have been implicated in various roles in plaque biofilm formation and dental caries pathogenesis, particularly GtfB and GtfC [Tanzer et al., 1985; Yamashita et al., 1993; Venkitaraman et al., 1995; Tamesada et al., 2004; Koo et al., 2010b; Bowen and Koo, 2011]. These enzymes are found in solution in saliva as well as bound to the salivary pellicle formed on the tooth surface or to microbial surfaces in their active forms. These enzymes can even be found bound to non-Gtf-producing oral bacteria [McCabe and Donkersloot, 1977; Rölla et al., 1983; Schilling and Bowen, 1988; Venkitaraman et al., 1995; Vacca-Smith and Bowen, 1998].

The glucans in the pellicle, which are produced by surface-adsorbed Gtfs (primarily GtfC followed by GtfB), provide avid binding sites that promote adherence of larger numbers of S. mutans and S. sobrinus as these organisms bind with greater adhesion strength to the in situ glucans than to saliva-coated apatitic (sHA) surfaces [Schilling and Bowen, 1992; Venkitaraman et al., 1995; Cross et al., 2007]. The binding of these organisms to glucans formed in situ is mediated by the presence of cell-associated Gtf enzymes and non-Gtf glucan-binding proteins (Gbps) such as GbpC [Banas and Vickerman, 2003] (fig. 1). Furthermore, it is apparent that glucan synthesized in situ by Gtfs in pellicle provides enhanced binding for several oral microorganisms [Bowen and Koo, 2011]. Concomitantly, glucan formed by Gtf adsorbed on the surface of microbial cells (primarily GtfB) enhances binding between bacterial cells, thus promoting the initial clustering of cells and the subsequent formation of microcolonies [Koo et al., 2010b]. These glucan-mediated processes increase the accumulation of cells tightly adherent to the surface, and enhance the cohesiveness and mechanical stability of the biofilms, allowing them to persist on tooth enamel for prolonged periods [Bowen and Koo, 2011]. Thus, the Gtfs (particularly GtfB and GtfC), in addition to Gbps, may be ideal targets for therapeutic intervention aiming at preventing plaque formation and dental caries.

As shown in table 1, the polyphenolic compounds have been extensively studied because of their ability to reduce glucan synthesis, although most studies use crude preparations of Gtfs in solution. Among these compounds, various polyphenols from the leaves of C. sinensis (extracts of green or oolong tea), propolis, cacao bean husk, cranberry and often traditional plants (Serindeia warnecki, Azadirachta indica, R. gardneriana and M. chinensis) exhibit inhibitory effects against glucan synthesis, particularly water-insoluble glucan. The biologically active compounds range from low-molecular-weight (LMW) flavonoids (e.g. in propolis), benzophenones, epigallocatechin/epicatechin gallate (e.g. in green tea) over smaller proanthocyanidin oligomers (e.g. in cranberries) to high-molecular-weight (HMW) polymeric polyphenols (e.g. in oolong tea, cacao and traditional medicinal plants) [Wolinsky and Sote, 1984; Wu-Yuan et al., 1988; Hattori et al., 1990; Otake et al., 1991; Nakahara et al., 1993; Hamada et al., 1996; Wolinsky et al., 1996; Hamilton-Miller, 2001; Osawa et al., 2001; Koo et al., 2002a, b, 2010a–c; Matsumoto et al., 2003; Murata et al., 2008].

According to Yanagida et al. [2000], the HMW plant polyphenols that display strong anti-Gtf activity have a common structural feature that is shared with catechin-based oligomeric forms (condensed tannins) and/or gallate ester form compounds (hydrolysable tannins). HMW polymeric proanthocyanidins (PAC), primarily composed of epicatechin units linked by C-4β and C-8 bonds, were identified as the major biologically active compounds in cocoa extracts [Osawa et al., 2001]. Furthermore, it appears that these inhibitory effects are enhanced with increasing polymerization of monomeric polyphenol [Hamada et al., 1996], which may be attributed to nonspecific binding and precipitation of the enzymes in solution (e.g. gallotannins) [Wu-Yuan et al., 1988]. However, these HMW compounds may have site-specific actions, as reported by Matsumoto et al. [2003], whose research has demonstrated that oolong tea fractions rich in polymeric polyphenols noncompetitively reduce glucan synthesis by targeting the glucan-binding domain of S. mutans-derived GtfB and GtfD in solution.

Furthermore, the type of interflavan linkage may also play a critical role in the anti-Gtf activity of polyphenols. Cranberry PAC dimers containing the A-type double interflavan bond (which confers structural rigidity to the molecule) display significant inhibitory activity [Gregoire et al., 2007; Koo et al., 2010a]. The B-type dimers derived from apple have a single linkage and have not been demonstrated to possess any such activity [Yanagida et al., 2000]. On the other hand, LMW apigenin and 7-epiclusianione appear to be effective noncompetitive inhibitors of GtfB and GtfC [Koo et al., 2002c; Murata et al., 2008]. The position of hydroxyl (OH) groups and the presence of a double bond between C2 and C3 in the flavone or flavonol molecular structure may provide a site for nucleophilic addition, which could disrupt the enzymatic activity of Gtfs [Koo et al., 2002c].

The effects of natural products on surface-adsorbed Gtfs have received limited attention despite their clinical importance (see Problems and Limitations section). Ethanolic extracts of Apis mellifera propolis (a resinous beehive product) and R. gardneriana displayed inhibitory effects against surface-adsorbed GtfB and GtfC, which was attributed to the presence of LMW inhibitors such as flavonols (e.g. kaempferol and myricetin), flavones (e.g. apigenin) and benzophenones (e.g. 7-epiclusianione) [Koo et al., 2002c; Murata et al., 2008]. It has recently been shown that specific A-type proanthocyanidin oligomers (from dimers to dodecamers) in an aqueous extract of cranberry effectively diminished the synthesis of insoluble glucans by surface-adsorbed GtfB [Gregoire et al., 2007; Koo et al., 2010a]. The presence of an A-type interflavan linkage and a degree of polymerization that ranged from 4 to 12 was considered optimal for the effective inhibition of the surface-adsorbed enzyme [Gregoire et al., 2007; Koo et al., 2010a]. In addition, the presence of apigenin (and other bioactive flavonoids) also decreased the expression of gtfB and gtfC (but not gtfD) in S. mutans growing planktonically or as a biofilm, all without affecting bacterial growth or culture pH [Koo et al., 2006]. However, little is known about the exact molecular mechanisms by which these naturally occurring molecules inhibit enzyme activity and gtf gene expression.

Inhibition of Bacterial Adherence

As shown in table 1, some natural products also inhibit bacterial adherence to hard surfaces (glass or hydroxyapatite) and/or cause bacterial aggregation. HMW polyphenols (from green tea, oolong tea, grape, cranberry and pine bark), chitosans and 1-deoxynojirimycin (isolated from Morus alba) have been shown to inhibit sucrose-dependent (EPS-mediated) and independent (pellicle-mediated) adherence of mutans streptococci [Tarsi et al., 1997; Steinberg et al., 2004; Koo et al., 2005; Furiga et al., 2008; Islam et al., 2008]. However, most of these studies were conducted when test agents were mixed with planktonic organisms, making data interpretation difficult due to overlapping biological activities (e.g. effects on growth, aggregation and Gtf activity). In general, the inhibition of sucrose-dependent adherence is associated with a reduction in water-insoluble glucan synthesis, which could decrease the accumulation of cariogenic bacteria and further biofilm development [Koo et al., 2005; Furiga et al., 2008; Islam et al., 2008].

Antimicrobial Activity

Despite early efforts to identify the putative antimicrobial agents from natural products, the precise mechanisms of action and effectiveness are still largely unknown owing to the limitations of the assays used to measure inhibition of growth (e.g. MIC and MBC determination). Such approaches to assess the antimicrobial activity of a possible anticaries agent are problematic for a number of reasons. First, bacteria in the oral cavity would never be exposed to a static (constant) concentration of an exogenously introduced antimicrobial agent over a 24-hour period. Unless a compound has an outstanding substantivity, microorganisms in the oral cavity are unlikely to be exposed to high concentrations of an antimicrobial for more than 30 s to a couple of minutes. Second, bacteria in classical MIC/MBC assays are in suspension, whereas the oral bacteria associated with caries are enmeshed in the plaque biofilm matrix. There is a plethora of evidence showing that biofilm cells are more resistant to antimicrobials than cells in suspension. Thus, describing antibacterial activity against planktonic oral bacteria with constant exposure to an agent at a high concentration over a 24-hour period is hardly relevant to how oral bacteria would respond in the mouth. Nevertheless, determination of MIC and MBC can still be useful when employed in high-throughput bioscreening techniques.

Despite incomplete information concerning their mechanisms of action and a lack of studies to evaluate treatment in a more clinically relevant biofilm model system, many natural biocidal agents have been incorporated in oral health care products. For example, extracts of miswak, tea tree oils, essential oils, peppermint and green tea have been added to mouthrinses and toothpastes to enhance their overall antimicrobial activities [Allaker and Douglas, 2009]. Furthermore, the biological relevance of agents displaying effects on bacterium growth needs to be interpreted with caution because antimicrobial activity does not necessarily correlate with clinical antiplaque effects, and there is no direct association with cariostatic activity [Scheie, 1989; Twetman, 2004].

Inhibition of EPS Synthesis

Following recognition of the importance of Gtfs in the pathogenesis of dental caries, specific inhibition of a proven virulence factor became an attractive and potentially highly selective approach to preventing plaque-related diseases. However, much of the work in this area has focused largely on enzymatic activity in solution using crude preparations, all without regard to the importance of the enzymes in the pellicle and kinetic differences among individual Gtfs. The development of dental caries can be attributed to events that occur at the tooth pellicle-plaque interface, and enzymatically active Gtfs are present in the pellicle and on bacterial surfaces. The identification of inhibitors that target these adsorbed enzymes is an attractive possibility for the development of new anticaries therapies. It is also noteworthy that surface-adsorbed Gtfs display an increased resistance to the most common inhibiting agents compared with solution phase enzymes, which includes the currently available antiplaque mouthrinses [Vacca-Smith and Bowen, 1998; Wunder and Bowen, 1999; Koo et al., 2002a]. The reason for this phenomenon is unclear, although it may be related to conformational changes that these enzymes undergo during adsorption, which can alter their enzymatic kinetics and physicochemical properties [Schilling and Bowen, 1988; Venkitaraman et al., 1995; Bowen and Koo, 2011].

Clearly, agents that inhibit enzymes in solution may be without effect when the enzymes are adsorbed to a surface. Our studies have shown that many natural products and their bioactive constituents display partial inhibition of GtfB and GtfC in solution, but only a few are potent inhibitors (>90% inhibition), and even fewer disrupt the activity of surface-adsorbed enzymes. In general, we have observed that effective surface-adsorbed GtfB and GtfC inhibitors also disrupt the EPS content in biofilms, reducing the biomass and modulating cariogenicity in vivo [Koo et al., 2003, 2005, 2010a; Koo and Jeon, 2009]. Thus, we would recommend that inhibitors of Gtfs should at minimum be as effective in inactivating surface-adsorbed enzymes as they are in inhibiting these enzymes in solution before any conclusions can be drawn concerning the disruptive effects a natural product may have on glucan synthesis.

Despite evidence that several natural products may have the potential to modulate the pathogenesis of dental caries, few studies have actually been conducted in vivo (e.g. using rodent models of dental caries) and even fewer have been evaluated at the clinical level (table 1). Here, we review some of the available, although limited, in vivo and clinical data. Some studies have shown that extracts of green tea or oolong tea enriched with polyphenols significantly reduce plaque accumulation and/or the development of carious lesions on the sulcal and/or smooth tooth surfaces of rats infected with S. mutans or S. sobrinus when delivered by drinking water or diet [Rosen et al., 1984; Otake et al., 1991; Ooshima et al., 1993]. The cariostatic effects of these agents may be related to the presence of (–)-epigallocatechin, (–)-epigallocatechin gallate and HMW polymeric polyphenols which display antibacterial and/or anti-Gtf activity [Otake et al., 1991; Ooshima et al., 1993].

Topical applications of chemically characterized propolis extracts have also been shown to be highly effective in reducing the incidence and severity of smooth surface and sulcal caries in rats [Ikeno et al., 1991; Koo et al., 1999; Hayacibara et al., 2005; Duarte et al., 2006b]. However, the cariostatic effects of propolis are highly variable depending on its chemical composition and geographical origin [Koo et al., 1999]. The biological activity is associated in part with the presence of flavonoids and terpenoids, such as apigenin and tt-farnesol, that display anti-GtfB/GtfC activity and inhibit acidogenicity and/or aciduricity in S. mutans [Koo et al., 2002c, 2003; Koo and Jeon, 2009]. Furthermore, apigenin and tt-farnesol (alone or in combination) significantly reduce caries development in rats with minimal effects on the population of viable cells in the plaque [Koo et al., 2003]. Topical applications of the PAC-rich fractions of cranberry extracts were recently demonstrated as having cariostatic effects in vivo. This may be attributed to the inhibition of bacterial glycolysis and glucan-mediated processes involved in biofilm formation, which appears to be exacted by specific bioactive A-type dimers and oligomers present in the PAC extract [Koo et al., 2010a].

Clinical studies evaluating anticaries effects are virtually nonexistent. However, some studies have demonstrated inhibitory effects on salivary levels of cariogenic bacteria (mutans streptococci and lactobacilli) and on plaque accumulation [Koo et al., 2002a; Matsumoto et al., 2004; Pai et al., 2004; Menezes et al., 2006]. For example, a mouthrinse containing propolis extracts (3% w/v) effectively reduced the accumulation and insoluble polysaccharide content of dental plaque (44.7 and 61.7% reduction, respectively, vs. placebo mouthrinse) [Koo et al., 2002a]. In the same context, oolong tea-based mouthwashes (0.05% w/v extract) reduced the mean plaque index by 21% [Ooshima et al., 1994]. Despite limited investigations in vivo, the available evidence indicates that there is potential for the discovery of novel and effective anticaries/antiplaque therapy using natural products. There is a clear need to conduct additional studies to evaluate the clinical efficacy and safety of these substances [Groppo et al., 2008].

Contemporary drug discovery is based on rapid screening of synthetic or naturally occurring molecules for their ability to affect specific pharmacological target(s), often using high-throughput screening, as reviewed in Koehn and Carter [2005] and Schmidt et al. [2007]. The complexity of crude or semipure natural extracts and the chemical nature of the components found in them are major challenges in discovering new bioactives. Furthermore, several rounds of purification and additional bioassays are required for the identification and isolation of the active components of an exceptionally complex matrix, which is laborious, technically challenging and time-consuming. Then, there is the need for structural elucidation and molecular formula determination, which is technically feasible using the current spectroscopic techniques, albeit difficult. Purification is a rate-limiting step because separation methods must be designed to yield sufficient quantities of the pure bioactive molecule from the original natural source. The latter highlights one of the fundamental concerns in natural product drug discovery: the limited availability of chemically characterized natural supplies.

Furthermore, the chemical composition of the naturally derived products depends on geographic location, seasonal effects and biological diversity. For example, there is a diverse range of Brazilian propolis, a beehive product from A. mellifera honeybees, as more than 13 types of propolis have been classified based on physicochemical characteristics, botanical origin and bioactivity. This variation was found to be associated with a number of factors, including (i) the biodiversity in Brazil, which is at least partially dictated by the geographical location, (ii) seasonal changes that affect plant growth and availability to the honeybees, and (iii) genetic variation in A. mellifera bees [Park et al., 1998, 2002; Bankova, 2005; Salatino et al., 2005; Silva et al., 2008; Castro et al., 2009a].

Biofilm Research

The effects of the natural products on bacterial growth, metabolism, adherence and polysaccharide synthesis clearly indicate that such agents would have an effect on initial formation and further accumulation of biofilm on apatitic surfaces. Surprisingly, very few studies have examined the effects of such products on biofilms related to caries such as those formed by mutans streptococci or other biofilm-forming cariogenic microorganisms [Koo et al., 2003, 2005, 2010a; Xie et al., 2008; Islam et al., 2009; Jeon et al., 2009; Cheng et al., 2011]. Thus, without further in vitro assessment of the antibiofilm activity of such agents in a more clinically relevant (brief exposure) model system, it is not safe to postulate their anticaries potential.

Many studies on natural products in the field of caries research have offered incomplete information because they lack basic chemical characterizations of the extracts or their compositional profiles and/or identification of putative active compound(s). Unfortunately, most biological assays commonly used are focused on the growth and metabolism of planktonic cells, and these cells are exposed for prolonged and inappropriate periods of time, considering biofilm development and treatment regimens in the context of dental caries disease.

It is evident that research involving natural products is complex. In conducting such studies, it is important to consider whether there is (i) a reliable and reproducible source of collection of and purification from the natural product, (ii) expertise in analytical and medicinal chemistry, and (iii) use of clinically relevant and feasible models for bioactivity evaluation. Therefore, a stepwise research plan in a multidisciplinary fashion is required, which may involve at least 6 major steps or phases: (1) discovery – finding natural sources and conducting bioscreens; (2) characterization of bioactives – identification, isolation and chemicostructural elucidation; (3) validation of biological activity – in vitro and in vivo studies using clinically relevant models as well as determination of mechanisms of action; (4) toxicology studies; (5) clinical studies of efficacy and safety (approval by FDA in the USA, or similar entities elsewhere); and (6) product development and commercialization. In the case of multicomponent botanical therapeutics (MCBT), which is a mixture of interacting compounds produced by plants, the extract should be chemically characterized and standardized before being evaluated using in vitro and in vivo models [Schmidt et al., 2007].

Here, we suggest a research model for initial evaluation (steps 1–3) of natural products in caries research based on our more than 15 years of experience. Steps 4–6 will not be discussed here as these require the highly specialized technical expertise of medicinal chemists, clinical scientists and toxicologists, which may also include industrial partnerships. A comprehensive program ranging from basic laboratory studies to in vivo investigations in animals is offered, which could aid in the development of new chemotherapeutic anticaries approaches for clinical testing. Our suggested strategy is diagramed in figure 2.

Fig. 2

Overview of the Suggested Model for Discovery of Naturally Occurring Anti-Caries Candidates.

Fig. 2

Overview of the Suggested Model for Discovery of Naturally Occurring Anti-Caries Candidates.

Close modal

Step 1: Discovery Phase1

Finding Natural Sources

Natural products can be selected and obtained via a number of approaches including but not limited to: (i) natural substances used in traditional medicine, (ii) systematic collection of a biodiverse set of natural products (plants, microbes or marine organisms), and/or (iii) selection of species of plants or other organisms (microbes and marine) based on their phylogenetic relationship to those known to produce bioactive compound(s) [Jones and Kinghorn, 2006].

The local micro- and macroenvironment, plant diversity, climate, geographic location and even seasonal variation (e.g. time of collection) significantly influence the content of bioactive compounds in natural products, which can dramatically influence the biological activity and potency of the samples [Sforcin et al., 2000; Bankova, 2005; Castro et al., 2007; Schmidt et al., 2007]. Finding a reliable, reproducible and unadulterated natural product source is a major hurdle both in medicine and dentistry. Thus, it is critical to work closely with local or native plant taxonomists and research groups to obtain representative samples according to collection period and site as well as identification of the plants or other natural sources. After collection, the samples should be stored at –20 or –80°C to prevent any degradation or enzymatic changes. The natural products must be precisely identified and the authenticity must be confirmed by a taxonomist. Dried specimens should also be available at any time as reference specimens that represent the natural products that were collected and used for the preparation of extracts [Kaufman et al., 1999].

Once a potential bioactive natural product is identified, the source material (e.g. plant or fruits) could be cultivated under defined environmental conditions to control climate and nutrition. DNA fingerprinting may also be used to evaluate and produce highly standardized active extracts. For example, optimized and chemically characterized extracts of Artemisia dracunculus (Russian tarragon) and Vaccinium macrocarpon (American cranberry) have been successfully obtained using such cultivation processes [Schmidt et al., 2007; Koo et al., 2010a].

Biological Screening

The initial screening of natural substances should be simple and designed to rapidly evaluate the potential bioactivity of the whole or crude extracts. Usually water, ethanol or an aqueous ethanol-based solvent is used to prepare plant or plant-derived extracts (e.g. propolis). The selection of biological targets is critical to the design of medium to high-throughput screens, which should be technically feasible, reproducible and cost effective. In order to identify natural products with potential anticaries properties, we would focus on three assays: (i) extracellular glucan synthesis by GtfB and GtfC in solution and adsorbed onto sHA surfaces, (ii) glycolytic pH drop assays for S. mutans or other cariogenic organisms to test acidogenicity and aciduricity, and (iii) bacterial growth rate over time using high-throughput systems such as the Bioscreen C. These assays are simple and affordable and can be rapidly performed with multiple replicates, all the while addressing established virulence traits associated with the pathogenesis of dental caries. The bioscreen step would assist in the identification of pharmacologically active natural products that could be further characterized through fractionation and purification steps.

Step 2: Bioactive Characterization Phase

The chemical composition of a bioactive crude extract can be examined using standard spectroscopic procedures such as high-performance liquid chromatography (HPLC) and gas chromatography/mass spectrometry (GC/MS). Promising extracts could then be subjected to a bioassay-guided fractionation and purification process, which allows for localization of the bioactivity and target-directed isolation of the compounds [Koehn and Carter, 2005]. Although new technologies are available for online bioassays with real-time fractionation and simultaneous identification/structure determination, they are still costly and demand special expertise for the immobilization of targets.

Identification, Isolation and Chemicostructural Elucidation

The selected active extract can be fractionated by several methods using chemical gradients based on polarity and column chromatography. Analysis of the chemical composition of the crude extract can provide some guidance on the selection of solvents, using polarity and relative biohazard ranking as a guide. We have used dry (e.g. silica gel) or liquid (e.g. Sephadex LH-20) chromatographic columns for the fractionation of various natural products from plant to food extracts [Hayacibara et al., 2005; Duarte et al., 2006a; Koo et al., 2010b]. The fractionation step allows for the initial separation of the various compounds from the crude mixture, and subsequent identification of the most biologically active fraction [Gunatilaka et al., 2001].

The active fraction can then be subjected to a series of chromatographic-spectroscopic methods including semipreparative HPLC to isolate and purify individual molecules (both known and unknown) that are present in the semipure mixture. Concomitantly, the isolated compounds need to be identified and structurally characterized, and their purity should be assessed. The following methods may be used to identify and determine the chemical structures based on chromatography (HPLC and GC), molecular weight (MS, X-ray, infrared and ultraviolet) and physical characteristics (nuclear magnetic resonance, NMR) among others (see details in Koehn and Carter [2005] and Castro et al. [2009a]). Advances in NMR, MS, HPLC and matrix-assisted laser desorption/ionization time-of-flight technologies and the ability to use them in tandem with one another (e.g. HPLC-NMR-MS) greatly enhances the isolation, analysis and structural determination of the compounds from complex natural product extracts. Although the determination of complex structures is technically challenging, it is no longer a major impediment to the contemporary drug discovery process. For example, individual LMW and HMW flavonoids can be isolated and characterized from complex mixtures, such as in cranberry extracts, in as little as a few weeks [Gregoire et al., 2007; Koo et al., 2010a]. Nevertheless, the choice of which one should be used depends on individual factors including the advantages and disadvantages of each method and the physicochemical characteristics of the natural substance(s). Subsequently, partnership with analytical chemists may be required [Koehn and Carter, 2005; Verpoorte et al., 2005; Castro et al., 2009a]. Thus, a complex and multidisciplinary approach is required, and will not be discussed further in this review.

Each isolated compound and the selected fraction should be subjected to dose-response studies using the assays described in the bioscreening step. We would draw particular attention to surface-adsorbed GtfB/C activity and acidogenicity/aciduricity using glycolytic pH drop assays. It would be expected that the isolated compound is more potent than the semipure fraction or the original crude extract. However, individual compounds frequently are not as effective as the active fractions or crude extract. This occurs because the biological activity of the natural whole product is a result of the activity of synergistic or additive interactions of different compounds in the mixture rather than of a single highly active molecule [Castro et al., 2009b]. Thus, at this stage a decision should be made about whether further analysis will be conducted using the individual compound (single-molecule type) or the semipure chemically characterized mixture (multi-component botanical therapeutics (MCBT) type).

Step 3: Biological Activity Validation Phase

The biological activity of selected natural agents should be confirmed by assays that take into account the pathophysiological aspects of caries disease (fig. 1), using clinically relevant, feasible and reproducible models. Laboratory studies should focus primarily on biofilms formed on hydroxyapatite (a surrogate for tooth enamel) or enamel surfaces that are placed in a vertical position and coated with a salivary pellicle [Koo et al., 2005, 2010b]. The test agents should be topically applied for a relatively short period of time (30–60 s) and then ideally would be removed from the system, allowing for a more tightly controlled substance exposure time, as these conditions would be most similar to those experienced in vivo (e.g. brief exposure of dental plaque to exogenously introduced mouthrinses) [Guggenheim et al., 2001; Koo et al., 2003]. Shear forces may also be applied to dislodge loosely bound bacteria and/or excess agents after treatment. This could be simply accomplished by swirling and dip-washing the biofilms during culture medium changes and after topical applications [Guggenheim et al., 2001; Koo et al., 2003]. Also, the presence of saliva in the system is particularly important, considering that salivary proteins, especially proline-rich proteins, are known to bind polyphenols [Bennick, 2002].

We suggest a three-stage process which utilizes in vitro mono- and mixed-species biofilms and an in vivo (rodent) model of dental caries to further evaluate the biological activity of the selected agents and to identify potential new anticaries therapies.

Stage 1

Initially, the previously selected active substances (pure individual compounds or fractions) should be tested using the simplest model system: monospecies biofilms. These biofilms are formed on sHA disks or enamel slabs placed vertically in 24-well plates [Koo et al., 2005, 2010b; Lemos et al., 2010; Klein et al., 2011]. A monospecies biofilm system does not reflect the complex microbial community found in coronal dental plaque, although this model incorporates specific elements associated with the formation of cariogenic biofilms (e.g. the presence of a glucan-rich matrix and culture of a proven virulent organism such as S. mutans UA159). This biofilm system is simple and cost effective, allowing for multiple replicates of a single experiment to minimize variability and allow for straightforward topical applications of the agent. Furthermore, biofilms using a single organism are advantageous as they allow for determination of the mechanisms of action of selected test agents and their overall effect on microbial physiology and the transcriptional responses to treatment. This model can be used to focus on glucan-mediated and other metabolic processes involved in biofilm matrix formation and acidogenicity.

In this system, biofilms are exposed to the selected agents twice daily (60-second exposure, each time followed by removal of the excess agent) until the end of the experimental period (usually 5 days). After treatments, the biofilms are either homogenized by sonication or kept intact for multiple biochemical assays. Homogenized biofilm suspensions can be used for (i) determination of biomass (dry weight), (ii) determination of the viable populations, (iii) total protein, and (iv) total polysaccharide content (e.g. EPS glucans and fructans, and IPS) [Koo et al., 2003]. The physiological response of intact biofilms can be analyzed using glycolytic pH drop, acid-killing, proton permeability and F-ATPase activity assays [Koo et al., 2003; Duarte et al., 2006a; Gregoire et al., 2007; Jeon et al., 2009; Lemos et al., 2010], while the spatial distribution/ratio of EPS to bacteria biomass can be assessed using confocal fluorescence imaging [Xiao and Koo, 2009; Koo et al., 2010b]. The surface of attachment (sHA or enamel slabs) can be examined for demineralization and structural changes using established techniques such as determination of microhardness, laser-induced fluorescence and surface topography analysis [Zero, 1995; Aires et al., 2006]. Furthermore, the persistence of biological effects of topically applied agents after brief exposure (and removal of excess agent) may also be an indicator of substantivity, which is critical for developing novel chemotherapeutic approaches against oral diseases such as dental caries [Brecx, 1997]. Studies using a monospecies biofilm model yield valuable information that can be applied to complex ecological interactions, which may be evaluated using a multispecies biofilm method in vitro (stage 2) and in a rodent model of dental caries in vivo (stage 3). This step can be used to identify and select agents and their optimal concentrations which may be most effective in disrupting the formation of biofilms by mechanisms that target the major virulence attributes of a cariogenic organism (e.g. glucan production, acidogenicity and aciduricity).

Stage 2

In this phase, the effects of agents selected from stage 1 are evaluated using a multispecies biofilm model that follows the ecological plaque hypothesis [Marsh, 2003]. Our multispecies biofilm model was designed to mimic the formation of cariogenic biofilms in a simple and reproducible system that allows for the short-term exposure of replicate biofilms to selected substances (see details in Koo et al. [2010b]). In this model, we can examine whether test agents affect the EPS matrix development, acidogenicity and ecological interactions of S. mutans in the presence of other relevant oral species such as early colonizers including non-mutans streptococci and Actinomyces spp. Biofilm development can be assessed before and after changing a key environmental factor: the introduction and persistent availability of sucrose. This can be accomplished by altering the concentration and the availability of sucrose provided in the culture media. In addition, all the bioassays and methods described for the monospecies model can be employed here as well. This comprehensive analysis can provide a detailed evaluation of the effectiveness of selected agents, as well as identify potential anticaries candidates based on their ability to: (i) reduce biofilm biomass, (ii) inhibit the formation and establishment of the EPS matrix, (iii) impair acid stress survival mechanisms of S. mutans or other cariogenic organisms, (iv) disrupt the formation of an aciduric environment within the mixed microbial community of the biofilm, and/or (v) reduce demineralization of the apatitic/enamel surface. At this point, following completion of stages 1 and 2, the candidate(s) can be selected based on potency and the type of bioactive [single molecule(s) or MCBT], and can then be validated in vivo (stage 3). There are many other multispecies biofilm models [Fontana et al., 1996; Guggenheim et al., 2001; Wong and Sissons, 2001; Tang et al., 2003] including those using an even more complex, mixed microflora (including artificial mouth systems), which can of course be used in this step and may provide additional relevant information about ecological changes to the biofilms and other in vitro parameters, as discussed above.

Stage 3

In the final stage, we suggest evaluating the efficacy of agents using an established rodent model of dental caries [Bowen et al., 1988; Koo et al., 1999]. This in vivo model simulates more precisely the variables encountered in the oral cavity, such as exposure to salivary and cellular components as well as hydrodynamic and abrasive forces. The principles of bacterial adhesion (Streptococcus and Actinomyces spp.) to HA surfaces coated with rat saliva are similar to those observed with human saliva coated HA surfaces [Kopec and Bowen, 1995]. Our rat model allows the formation of cariogenic dental plaque on the tooth enamel surface of molars when the animals are provided a sucrose-rich diet (as would occur in humans). In addition, the rat harbors a complex and mixed oral flora even when infected with S. mutans or other cariogenic streptococci. More importantly, the model allows the precise examination of the incidence and severity of carious lesions on smooth, sulcal and interproximal surfaces simultaneously (and dynamically). We believe that there is no other model that can simulate such a range of variables found in the oral cavity and at the same time assess the complexity of the development of carious lesions in such a variety of surfaces. Properly designed and conducted studies using a rat caries model, despite its limitations, remain ethically sound and the most comprehensive and reproducible way of evaluating therapeutic agents preclinically. In alignment with stages 1 and 2, the test agents may be applied topically twice daily, and the impact of treatment may be assessed via the following: (i) determination of total aerobic and anaerobic cell populations recovered from animals’ plaque; (ii) determination of S. mutans (or other cariogenic strains of interest); (iii) the ratios of these populations, and (iv) caries scoring according to Larson’s modification of the Keyes system [Larson, 1981]. The scoring system assesses the incidence of dental caries on smooth, sulcal and interproximal surfaces, and the degree of severity of these carious lesions. The data may serve as an indicator of the efficacy of treatment under given experimental conditions which are designed to reflect the conditions likely to be experienced in vivo in the human mouth (e.g. brief exposure and clearance of the agents in the oral cavity). In this design, the animals are fed a sucrose-rich diet, which leads to development of carious lesions similar to that in humans. Thus, we can compare the abilities of various agents to prevent or reduce the initiation and progression of the disease by including positive controls known to be clinically effective in preventing dental caries (fluoride at 250 or 1,100 ppm) and plaque formation (chlorhexidine). Although our goal is to identify anticaries agents that are more effective than fluoride, we are also interested in agents that could be used as potential adjuvants to fluoride or other therapies (see later). We have already implemented this stepwise protocol to identify and evaluate specific bioactive molecules from cranberry and propolis, displaying potential anticaries effects [Koo et al., 2002c, 2003, 2005, 2010a; Duarte et al., 2006a; Gregoire et al., 2007].

The in vivo model may also allow for initial evaluation of the potential toxic effects of topical applications of test agents by monitoring daily food intake and weight gain, and by performing histopathological evaluations of major organs and mucosal surfaces. Although limited, this information may be helpful in designing future toxicological studies, including toxicopharmacokinetics, genotoxicity and carcinogenicity. Traditional in vitro studies to measure cytotoxicity and mutagenicity assays under high-throughput conditions may be combined with functional genomics and computational biology to enhance the ability to predict toxic effects of chemical agents [Committee on Toxicity Testing and Assessment of Environmental Agents, 2007]. However, such studies require a team of experts that can carefully select and perform assays.

Determination of Mechanisms of Action and Combination with Fluoride

Once a potential candidate is selected, it is critical to elucidate the molecular mechanisms of action by which these compounds disrupt the ability of cariogenic bacteria to form EPS and acids, and survive stress and an acidic environment. Understanding these molecular mechanisms may allow for the enhancement of their therapeutic effects. We suggest characterizing the impact of the selected agents on the expression of genes and their encoded proteins required for (i) EPS-rich matrix synthesis, (ii) acid tolerance response/stress survival mechanisms, (iii) enzymatic activity of Gtfs, (iv) sugar uptake systems, and (v) glycolytic enzymes, which are likely to modulate the pathogenesis of caries disease. We are in the process of developing protocols that may be used to determine the molecular mechanisms of action using a combination of standard biochemical assays and whole transcriptome/proteome profiling technologies (e.g. microarrays, RNA-Seq and MudPIT). Some of these protocols have recently been combined in an analytical toolbox for biofilm analyses [Klein et al., 2011]. This approach could then be used to identify the precise pharmacological targets of such agents in order to improve their effectiveness. Agents could then be tailored, including fluoride or other bioactive agents, to impact separate and/or complementary targets associated with EPS-rich matrix formation as well as acidogenicity and aciduricity, which may provide maximum therapeutic activity.

Fluoride in various preparations is arguably the mainstay of caries prevention. However, current applications of fluoride (i) offer incomplete protection against caries, and (ii) do not effectively address the infectious aspect of the disease. Nevertheless, fluoride does to some extent impede insoluble glucan synthesis, acid production and acid tolerance of cariogenic streptococci at micromolar levels found in the plaque matrix [Marquis et al., 2003; van Loveren, 2004]. Thus, it is promising that a combination of natural agent(s) acting in concert with fluoride may enhance the overall inhibitory effects that fluoride exerts on the virulence attributes of cariogenic bacteria. This may represent a superior approach to the prevention and treatment of dental caries, as demonstrated and reviewed by Koo et al. [2005, 2008].

Natural products remain a largely unexplored source of effective antibiofilm molecules of potentially low toxicity that could be used in alternative or adjunctive anticaries therapies. The emergence of powerful new analytical technologies and advances in ‘-omics’-based knowledge and in (bio)synthetic chemistry has opened the door to a new era in the development of novel antiplaque/anticaries therapies using naturally occurring agents. It is currently possible to obtain large quantities of purified protein targets for inhibitor screening (e.g. GtfB and GtfC). Furthermore, new separation and structure determination technologies have increased the plausibility of screening mixtures of structurally complex molecules. More importantly, advances in plant genomics, metabolic engineering and biosynthetic chemistry can now provide reproducible and optimized sources for the isolation and/or production of bioactive agents. These natural molecules can also be used as a template for the generation of synthetic and semisynthetic ligands using in silico approaches [Clardy and Walsh, 2004; Koehn and Carter, 2005; Dürig et al., 2010]. Although natural products from traditional medicine or plant-derived foods are generally not expected to have acute toxicological effects, comprehensive safety studies, especially following brief-exposure topical application of these agents, need to be evaluated prior to clinical trials [Groppo et al., 2008; Wu et al., 2008]. In addition, innovative delivery systems and improved solubilization methods would greatly enhance the applicability of natural products as anticaries agents.

Finally, natural agents can be used alone as single molecules, as MCBT and/or in combination with other agents for enhanced therapeutic effects. The use of natural agents to enhance the cariostatic properties of fluoride (without increasing the concentration of exposure) could be a promising and potent novel anticaries strategy [Koo and Jeon, 2009]. Clearly, there is great potential for the discovery of therapeutically relevant compounds from nature. However, careful experimental designs using multidisciplinary approaches in conjunction with the standardization and characterization of natural products are critical for the successful development of novel and useful anticaries chemotherapy. The implementation of standardized and stepwise cross-disciplinary approaches, as outlined here, would help to advance this field of investigation in caries research.

The authors’ research in this area was supported by grants from the National Institute for Dental and Craniofacial Research (NIDCR/NIH R01 DE016139 and DE018023), Brazilian Government Funding Agencies (CAPES/CNPq) and the Korean Ministry for Health, Welfare and Family Affairs (Korea Healthcare Technology R&D Project A084110).

The authors declare no conflict of interest is correlated to this paper.

1.
Aires CP, Tabchoury CP, del Bel Cury AA, Koo H, Cury JA: Effect of sucrose concentration on dental biofilm formed in situ and on enamel demineralization. Caries Res 2006;40:28–32.
2.
Allaker RP, Douglas CW: Novel anti-microbial therapies for dental plaque-related diseases. Int J Antimicrob Agents 2009;33:8–13.
3.
Almas K, Al-Zeid Z: The immediate antimicrobial effect of a toothbrush and miswak on cariogenic bacteria: a clinical study. J Contemp Dent Pract 2004;5:105–114.
4.
Axelsson P: Current role of pharmaceuticals in prevention of caries and periodontal disease. Int Dent J 1993;43:473–482.
5.
Bae K, Jun EJ, Lee SM, Paik DI, Kim JB: Effect of water-soluble reduced chitosan on Streptococcus mutans, plaque regrowth and biofilm vitality. Clin Oral Investig 2006;10:102–107.
6.
Bakkali F, Averbeck S, Averbeck D, Idaomar M: Biological effects of essential oils: a review. Food Chem Toxicol 2008;46:446–475.
7.
Bakri IM, Douglas CW: Inhibitory effect of garlic extract on oral bacteria. Arch Oral Biol 2005;50:645–651.
8.
Banas JA, Vickerman MM: Glucan-binding proteins of the oral streptococci. Crit Rev Oral Biol Med 2003;14:89–99.
9.
Bankova V: Chemical diversity of propolis and the problem of standardization. J Ethnopharmacol 2005;100:114–117.
10.
Beighton D: The complex oral microflora of high-risk individuals and groups and its role in the caries process. Community Dent Oral Epidemiol 2005;33:248–255.
11.
Bennick A: Interaction of plant polyphenols with salivary proteins. Crit Rev Oral Biol Med 2002;13:184–196.
12.
Bowen WH: Do we need to be concerned about dental caries in the coming millennium? Crit Rev Oral Biol Med 2002;13:126–131.
13.
Bowen WH, Koo H: Biology of Streptococcus mutans-derived glucosyltransferases: role in extracellular matrix formation of cariogenic biofilms. Caries Res 2011;45:69–86.
14.
Bowen WH, Pearson SK, Young DA: The effect of desalivation on coronal and root surface caries in rats. J Dent Res 1988;67:21–23.
15.
Brecx M: Strategies and agents in supragingival chemical plaque control. Periodontol 2000 1997;15:100–108.
16.
Brighenti FL, Luppens SB, Delbem AC, Deng DM, Hoogenkamp MA, Gaetti-Jardim E Jr, Dekker HL, Crielaard W, ten Cate JM: Effect of Psidium cattleianum leaf extract on Streptococcus mutans viability, protein expression and acid production. Caries Res 2008;42:148–154.
17.
Busscher HJ, Engels E, Dijkstra RJ, van der Mei HC: Influence of a chitosan on oral bacterial adhesion and growth in vitro. Eur J Oral Sci 2008;116:493–495.
18.
Cai S, Simionato MR, Mayer MP, Novo NF, Zelante F: Effects of subinhibitory concentrations of chemical agents on hydrophobicity and in vitro adherence of Streptococcus mutans and Streptococcus sanguis. Caries Res 1994;28:335–341.
19.
Castro ML, Cury JA, Rosalen PL, Alencar SM, Ikegaki M, Duarte S, Koo H: Própolis do sudeste e nordeste do Brasil: influência da sazonalidade na atividade antibacteriana e composição fenólica. Quim Nova 2007;30: DOI: 10.1590/S0100-40422007000700003.
20.
Castro ML, do Nascimento AM, Ikegaki M, Costa-Neto CM, Alencar SM, Rosalen PL: Identification of a bioactive compound isolated from Brazilian propolis type 6. Bioorg Med Chem 2009a;17:5332–5335.
21.
Castro ML, Vilela WR, Zauli RC, Ikegaki M, Rehder VL, Foglio MA, de Alencar SM, Rosalen PL: Bioassay guided purification of the antimicrobial fraction of a Brazilian propolis from Bahia state. BMC Complement Altern Med 2009b;9:25.
22.
Cheng L, Exterkate RA, Zhou X, Li J, ten Cate JM: Effect of Galla chinensis on growth and metabolism of microcosm biofilms. Caries Res 2011;45:87–92.
23.
Chinou I: Primary and secondary metabolites and their biological activity; in Waksmundzka-Hajnos M, Sherma J, Kowalska T (eds): Thin Layer Chromatography in Phytochemistry. New York, CRC, 2008, pp 59–76.
24.
Choi BK, Kim KY, Yoo YJ, Oh SJ, Choi JH, Kim CY: In vitro antimicrobial activity of a chitooligosaccharide mixture against Actinobacillus actinomycetemcomitans and Streptococcus mutans. Int J Antimicrob Agents 2001;18:553–557.
25.
Clardy J, Walsh C: Lessons from natural molecules. Nature 2004;432:829–837.
26.
Committee on Toxicity Testing and Assessment of Environmental Agents, National Research Council: Toxicity testing in the 21st century: a vision and a strategy. 2007. http://www.nap.edu/catalog/11970.html.
27.
Cowan MM: Plant products as antimicrobial agents. Clin Microbiol Rev 1999;12:564–582.
28.
Cross SE, Kreth J, Zhu L, Sullivan R, Shi W, Qi F, Gimzewski JK: Nanomechanical properties of glucans and associated cell-surface adhesion of Streptococcus mutans probed by atomic force microscopy under in situ conditions. Microbiology 2007;153:3124–3132.
29.
Darout IA, Albandar JM, Skaug N, Ali RW: Salivary microbiota levels in relation to periodontal status, experience of caries and miswak use in Sudanese adults. J Clin Periodontol 2002;29:411–420.
30.
Duarte S, Gregoire S, Singh AP, Vorsa N, Schaich K, Bowen WH, Koo H: Inhibitory effects of cranberry polyphenols on formation and acidogenicity of Streptococcus mutans biofilms. FEMS Microbiol Lett 2006a;257:50–56.
31.
Duarte S, Rosalen PL, Hayacibara MF, Cury JA, Bowen WH, Marquis RE, Rehder VL, Sartoratto A, Ikegaki M, Koo H: The influence of a novel propolis on mutans streptococci biofilms and caries development in rats. Arch Oral Biol 2006b;51:15–22.
32.
Dürig A, Kouskoumvekaki I, Vejborg RM, Klemm P: Chemoinformatics-assisted development of new anti-biofilm compounds. Appl Microbiol Biotechnol 2010;87:309–317.
33.
Dye BA, Tan S, Smith V, Lewis BG, Barker LK, Thornton-Evans G, Eke PI, Beltrán-Aguilar ED, Horowitz AM, Li CH: Trends in oral health status: United States, 1988–1994 and 1999–2004. Vital Health Stat 11 2007;248:1–92.
34.
Dzink JL, Socransky SS: Comparative in vitro activity of sanguinarine against oral microbial isolates. Antimicrob Agents Chemother 1985;27:663–665.
35.
Eley BM: Antibacterial agents in the control of supragingival plaque: a review. Br Dent J 1999;186:286–296.
36.
Fontana M, Dunipace AJ, Gregory RL, Noblitt TW, Li Y, Park KK, Stookey GK: An in vitro microbial model for studying secondary caries formation. Caries Res 1996;30:112–118.
37.
Fujiwara M, Hayashi Y, Ohara N: Inhibitory effect of water-soluble chitosan on growth of Streptococcus mutans. New Microbiol 2004;27:83–86.
38.
Furiga A, Lonvaud-Funel A, Dorignac G, Badet C: In vitro anti-bacterial and anti-adherence effects of natural polyphenolic compounds on oral bacteria. J Appl Microbiol 2008;105:1470–1476.
39.
Godowski KC: Antimicrobial action of sanguinarine. J Clin Dent 1989;1:96–101.
40.
Greenberg M, Dodds M, Tian M: Naturally occurring phenolic antibacterial compounds show effectiveness against oral bacteria by a quantitative structure-activity relationship study. J Agric Food Chem 2008;56:11151–11156.
41.
Gregoire S, Singh AP, Vorsa N, Koo H: Influence of cranberry phenolics on glucan synthesis by glucosyltransferases and Streptococcus mutans acidogenicity. J Appl Microbiol 2007;103:1960–1968.
42.
Groppo FC, de Cássia Bergamaschi C, Cogo K, Franz-Montan M, Motta RH, de Andrade ED: Use of phytotherapy in dentistry. Phytother Res 2008;22:993–998.
43.
Groppo FC, Ramacciato JC, Simões RP, Flório FM, Sartoratto A: Antimicrobial activity of garlic, tea tree oil, and chlorhexidine against oral microorganisms. Int Dent J 2002;52:433–437.
44.
Guggenheim B, Giertsen E, Schüpbach P, Shapiro S: Validation of an in vitro biofilm model of supragingival plaque. J Dent Res 2001;80:363–370.
45.
Gunatilaka AAL, Berger JM, Evans R, Miller JS, Wisse JH, Neddermann KM, Bursuker I, Kingston DGI: Isolation, synthesis, and structure-activity relationships of bioactive benzoquinones from Miconia lepidota from the Suriname rainforest. J Nat Prod 2001;64:2–5.
46.
Hamada S, Kontani M, Hosono H, Ono H, Tanaka T, Ooshima T, Mitsunaga T, Abe I: Peroxidase-catalyzed generation of catechin oligomers that inhibit glucosyltransferase from Streptococcus sobrinus. FEMS Microbiol Lett 1996;143:35–40.
47.
Hamilton-Miller JM: Anti-cariogenic properties of tea (Camellia sinensis). J Med Microbiol 2001;50:299–302.
48.
Hammer KA, Dry L, Johnson M, Michalak EM, Carson CF, Riley TV: Susceptibility of oral bacteria to Melaleuca alternifolia (tea tree) oil in vitro. Oral Microbiol Immunol 2003;18:389–392.
49.
Hattori M, Kusumoto IT, Namba T, Ishigami T, Hara Y: Effect of tea polyphenols on glucan synthesis by glucosyltransferase from Streptococcus mutans. Chem Pharm Bull 1990;38:717–720.
50.
Hayacibara MF, Koo H, Rosalen PL, Duarte S, Franco EM, Bowen WH, Ikegaki M, Cury JA: In vitro and in vivo effects of isolated fractions of Brazilian propolis on caries development. J Ethnopharmacol 2005;101:110–115.
51.
Hayashi Y, Ohara N, Ganno T, Yamaguchi K, Ishizaki T, Nakamura T, Sato M: Chewing chitosan-containing gum effectively inhibits the growth of cariogenic bacteria. Arch Oral Biol 2007;52:290–294.
52.
Hirasawa M, Shouji N, Neta T, Fukushima K, Takada K: Three kinds of antibacterial substances from Lentinus edodes (Berk) Sing (Shiitake, an edible mushroom). Int J Antimicrob Agents 1999;11:151–157.
53.
Hirasawa M, Takada K, Otake S: Inhibition of acid production in dental plaque bacteria by green tea catechins. Caries Res 2006;40:265–270.
54.
Hirschfeld I: The Toothbrush: Its Use and Abuse. A Treatise on Preventive Dentistry and Periodontia as Related to Dental Hygiene. New York, Dental Items of Interest Publishing, 1939.
55.
Hoover JN, To T: Efficacy of chlorhexidine and sanguinarine mouthrinses on selected salivary microflora. J Can Dent Assoc 1990;56:325–327.
56.
Ikeno K, Ikeno T, Miyazawa C: Effects of propolis on dental caries in rats. Caries Res 1991;25:347–351.
57.
Inoue Y, Shiraishi A, Hada T, Hirose K, Hamashima H, Shimada J: The antibacterial effects of terpene alcohols on Staphylococcus aureus and their mode of action. FEMS Microbiol Lett 2004;237:325–331.
58.
Islam B, Khan SN, Haque I, Alam M, Mushfiq M, Khan AU: Novel anti-adherence activity of mulberry leaves: inhibition of Streptococcus mutans biofilm by 1-deoxynojirimycin isolated from Morus alba. J Antimicrob Chemother 2008;62:751–757.
59.
Islam B, Khan SN, Naeem A, Sharma V, Khan AU: Novel effect of plant lectins on the inhibition of Streptococcus mutans biofilm formation on saliva-coated surface. J Appl Microbiol 2009;106:1682–1689.
60.
Jagtap AG, Karkera SG: Extract of Juglandaceae regia inhibits growth, in vitro adherence, acid production and aggregation of Streptococcus mutans. J Pharm Pharmacol 2000;52:235–242.
61.
Jeon JG, Klein MI, Xiao J, Gregoire S, Rosalen PL, Koo H: Influences of naturally occurring agents in combination with fluoride on gene expression and structural organization of Streptococcus mutans in biofilms. BMC Microbiol 2009;9:228.
62.
Jones WP, Kinghorn AD: Extraction of plant secondary metabolites; in Sarker SD, Latif Z, Gray AI (eds): Natural Products Isolation, ed 3. New Jersey, Humana, 2006, pp 324–351.
63.
Kaufman PB, Cseke LJ, Warber S, Duke JA, Brielmann HL: Natural Products from Plants. Boca Raton, CRC, 1999.
64.
Klein MI, Xiao J, Heydorn A, Koo H: An analytical tool-box for comprehensive biochemical, structural and transcriptome evaluation of oral biofilms mediated by mutans streptococci. J Vis Exp DOI: 10.3791/ 2512.
65.
Koehn FE, Carter GT: The evolving role of natural products in drug discovery. Nat Rev Drug Discov 2005;4:206–220.
66.
Koo H: Strategies to enhance the biological effects of fluoride on dental biofilms. Adv Dent Res 2008;20:17–21.
67.
Koo H, Cury JA, Rosalen PL, Ambrosano GM, Ikegaki M, Park YK: Effect of a mouthrinse containing selected propolis on 3-day dental plaque accumulation and polysaccharide formation. Caries Res 2002a;36:445–448.
68.
Koo H, Pearson SK, Scott-Anne K, Abranches J, Cury JA, Rosalen PL, Park YK, Marquis RE, Bowen WH: Effects of apigenin and tt-farnesol on glucosyltransferase activity, biofilm viability and caries development in rats. Oral Microbiol Immunol 2002b;17:337–343.
69.
Koo H, Rosalen PL, Cury JA, Park YK, Bowen WH: Effects of compounds found in propolis on Streptococcus mutans growth and on glucosyltransferase activity. Antimicrob Agents Chemother 2002c;46:1302–1309.
70.
Koo H, Hayacibara MF, Schobel BD, Cury JA, Rosalen PL, Park YK, Vacca-Smith AM, Bowen WH: Inhibition of Streptococcus mutans biofilm accumulation and polysaccharide production by apigenin and tt-farnesol. J Antimicrob Chemother 2003;52:782–789.
71.
Koo H, Jeon JG: Naturally occurring molecules as alternative therapeutic agents against cariogenic biofilms. Adv Dent Res 2009;21:63–68.
72.
Koo H, Rosalen PL, Cury JA, Park YK, Ikegaki M, Sattler A: Effect of Apis mellifera propolis from two Brazilian regions on caries development in desalivated rats. Caries Res 1999;33:393–400.
73.
Koo H, Schobel B, Scott-Anne K, Watson G, Bowen WH, Cury JA, Rosalen PL, Park YK: Apigenin and tt-farnesol with fluoride effects on S. mutans biofilms and dental caries. J Dent Res 2005;84:1016–1020.
74.
Koo H, Seils J, Abranches J, Burne RA, Bowen WH, Quivey RG Jr: Influence of apigenin on gtf gene expression in Streptococcus mutans UA159. Antimicrob Agents Chemother 2006;50:542–546.
75.
Koo H, Vacca Smith AM, Bowen WH, Rosalen PL, Cury JA, Park YK: Effects of Apis mellifera propolis on the activities of streptococcal glucosyltransferases in solution and adsorbed onto saliva-coated hydroxyapatite. Caries Res 2000;34:418–426.
76.
Koo H, Duarte S, Murata RM, Scott-Anne K, Gregoire S, Watson GE, Singh AP, Vorsa N: Influence of cranberry proanthocyanidins on formation of biofilms by Streptococcus mutans on saliva-coated apatitic surface and on dental caries development in vivo. Caries Res 2010a;44:116–126.
77.
Koo H, Xiao J, Klein MI, Jeon JG: Exopolysaccharides produced by Streptococcus mutans glucosyltransferases modulate the establishment of microcolonies within multispecies biofilms. J Bacteriol 2010b;192:3024–3032.
78.
Kopec LK, Bowen WH. Adherence of microorganisms to rat salivary pellicles. Caries Res 1995;29:507–512.
79.
Larson RM: Merits and modifications of scoring rat dental caries by Keyes’ method; in Tanzer JM (ed): Microbiology Abstracts. Animal Models in Cariology. Oxford, IRL, 1981, special suppl, pp 195–203.
80.
Lemos JA, Abranches J, Koo H, Marquis RE, Burne RA: Protocols to study the physiology of oral biofilms. Methods Mol Biol 2010;666:87–102.
81.
Lemos JA, Burne RA: A model of efficiency: stress tolerance by Streptococcus mutans. Microbiology 2008;154:3247–3255.
82.
Li XC, Cai L, Wu CD: Antimicrobial compounds from Ceanothus americanus against oral pathogens. Phytochemistry 1997;46:97–102.
83.
Linke HA, LeGeros RZ: Black tea extract and dental caries formation in hamsters. Int J Food Sci Nutr 2003;54:89–95.
84.
Loesche WJ: Role of Streptococcus mutans in human dental decay. Microbiol Rev 1986;50:353–380.
85.
Loesche WJ, Henry CA: Intracellular microbial polysaccharide production and dental caries in a Guatemalan Indian Village. Arch Oral Biol 1967;12:189–194.
86.
Marquis RE, Clock SA, Mota-Meira M: Fluoride and organic weak acids as modulators of microbial physiology. FEMS Microbiol Rev 2003;26:493–510.
87.
Marsh PD: Antimicrobial strategies in the prevention of dental caries. Caries Res 1993;27(suppl 1):72–76.
88.
Marsh PD: Are dental diseases examples of ecological catastrophes? Microbiology 2003;149:279–294.
89.
Matsumoto M, Hamada S, Ooshima T: Molecular analysis of the inhibitory effects of oolong tea polyphenols on glucan-binding domain of recombinant glucosyltransferases from Streptococcus mutans MT8148. FEMS Microbiol Lett 2003;228:73–80.
90.
Matsumoto M, Minami T, Sasaki H, Sobue S, Hamada S, Ooshima T: Inhibitory effects of oolong tea extract on caries-inducing properties of mutans streptococci. Caries Res 1999;33:441–445.
91.
Matsumoto M, Tsuji M, Okuda J, Sasaki H, Nakano K, Osawa K, Shimura S, Ooshima T: Inhibitory effects of cacao bean husk extract on plaque formation in vitro and in vivo. Eur J Oral Sci 2004;112:249–252.
92.
McCabe RM, Donkersloot JA: Adherence of Veillonella species mediated by extracellular glucosyltransferase from Streptococcus salivarius. Infect Immun 1977;18:726–734.
93.
Menezes SMS, Cordeiro LN, Viana GSB: Punica granatum (pomegranate) extract is active against dental plaque. J Herb Pharmacother 2006;6:79–92.
94.
Murata RM, Branco de Almeida LS, Yatsuda R, dos Santos MH, Nagem TJ, Rosalen PL, Koo H: Inhibitory effects of 7-epiclusianone on glucan synthesis, acidogenicity and biofilm formation by Streptococcus mutans. FEMS Microbiol Lett 2008;282:174–181.
95.
Nakahara K, Kawabata S, Ono H, Ogura K, Tanaka T, Ooshima T, Hamada S: Inhibitory effect of oolong tea polyphenols on glycosyltransferases of mutans streptococci. Appl Environ Microbiol 1993;59:968–973.
96.
National Institutes of Health: Diagnosis and management of dental caries throughout life. NIH Consens Statement 2001;18:1–30.
97.
Newman DJ, Cragg GM: Natural products as sources of new drugs over the last 25 years. J Nat Prod 2007;70:461–477.
98.
Ooshima T, Minami T, Aono W, Izumitani A, Sobue S, Fujiwara T, Kawabata S, Hamada S: Oolong tea polyphenols inhibit experimental dental caries in SPF rats infected with mutans streptococci. Caries Res 1993;27:124–129.
99.
Ooshima T, Minami T, Aono W, Tamura Y, Hamada S: Reduction of dental plaque deposition in humans by oolong tea extract. Caries Res 1994;28:146–149.
100.
Ooshima T, Minami T, Matsumoto M, Fujiwara T, Sobue S, Hamada S: Comparison of the cariostatic effects between regimens to administer oolong tea polyphenols in SPF rats. Caries Res 1998;32:75–80.
101.
Ooshima T, Osaka Y, Sasaki H, Osawa K, Yasuda H, Matsumura M, Sobue S, Matsumoto M: Caries inhibitory activity of cacao bean husk extract in in vitro and animal experiments. Arch Oral Biol 2000;45:639–645.
102.
Osawa K, Miyazaki K, Shimura S, Okuda J, Matsumoto M, Ooshima T: Identification of cariostatic substances in the cacao bean husk: their anti-glucosyltransferase and antibacterial activities. J Dent Res 2001;80:2000–2004.
103.
Otake S, Makimura M, Kuroki T, Nishihara Y, Hirasawa M: Anticaries effects of polyphenolic compounds from Japanese green tea. Caries Res 1991;25:438–443.
104.
Paes Leme AF, Koo H, Bellato CM, Bedi G, Cury JA: The role of sucrose in cariogenic dental biofilm formation: new insight. J Dent Res 2006;85:878–887.
105.
Pai MR, Acharya LD, Udupa N: Evaluation of antiplaque activity of Azadirachta indica leaf extract gel: a 6-week clinical study. J Ethnopharmacol 2004;90:99–103.
106.
Park YK, Alencar SM, Aguiar CL: Botanical origin and chemical composition of Brazilian propolis. J Agric Food Chem 2002;50:2502–2506.
107.
Park YK, Koo MH, Abreu JA, Ikegaki M, Cury JA, Rosalen PL: Antimicrobial activity of propolis on oral microorganisms. Curr Microbiol 1998;36:24–28.
108.
Quivey RG Jr, Kuhnert WL, Hahn K: Adaptation of oral streptococci to low pH. Adv Microb Physiol 2000;42:239–274.
109.
Rasooli I, Shayegh S, Taghizadeh M, Astaneh SD: Phytotherapeutic prevention of dental biofilm formation. Phytother Res 2008;22:1162–1167.
110.
Rölla G, Ciardi JE, Schultz AS: Adsorption of glucosyltransferase to saliva coated hydroxyapatite: possible mechanism for sucrose dependent bacterial colonization of teeth. Scand J Dent Res 1983;91:112–117.
111.
Rosen S, Elvin-Lewis M, Beck FM, Beck EX: Anticariogenic effects of tea in rats. J Dent Res 1984;63:658–660.
112.
Saeki Y, Kato T, Naito Y, Takazoe I, Okuda K: Inhibitory effects of funoran on the adherence and colonization of mutans streptococci. Caries Res 1996;30:119–125.
113.
Sakanaka S, Kim M, Taniguchi M, Yamamoto T: Antibacterial substances in Japanese green tea extract against Streptococcus mutans, a cariogenic bacterium. Agric Biol Chem 1989;53:2307–2311.
114.
Salatino A, Teixeira EW, Negri G, Message D: Origin and chemical variation of Brazilian propolis. Evid Based Complement Alternat Med 2005;2:33–38.
115.
Sasaki H, Matsumoto M, Tanaka T, Maeda M, Nakai M, Hamada S, Ooshima T: Antibacterial activity of polyphenol components in oolong tea extract against Streptococcus mutans. Caries Res 2004;38:2–8.
116.
Sato S, Yoshinuma N, Ito K, Tokumoto T, Takiguchi T, Suzuki Y, Murai S: The inhibitory effect of funoran and eucalyptus extract-containing chewing gum on plaque formation. J Oral Sci 1998;40:115–117.
117.
Scheie AA: Modes of action of currently known chemical anti-plaque agents other than chlorhexidine. J Dent Res 1989;68:1609–1616.
118.
Schilling KM, Bowen WH: The activity of glucosyltransferase adsorbed onto saliva-coated hydroxyapatite. J Dent Res 1988;67:2–8.
119.
Schilling KM, Bowen WH: Glucans synthesized in situ in experimental salivary pellicle function as specific binding sites for Streptococcus mutans. Infect Immun 1992;60:284–295.
120.
Schmidt BM, Ribnicky DM, Lipsky PE, Raskin I: Revisiting the ancient concept of botanical therapeutics. Nat Chem Biol 2007;3:360–366.
121.
Sforcin JM, Fernandes A Jr, Lopes CA, Bankova V, Funari SR: Seasonal effect on Brazilian propolis antibacterial activity. J Ethnopharmacol 2000;73:243–249.
122.
Shapiro S, Guggenheim B: The action of thymol on oral bacteria. Oral Microbiol Immunol 1995;10:241–246.
123.
Shapiro S, Meier A, Guggenheim B: The antimicrobial activity of essential oils and essential oil components towards oral bacteria. Oral Microbiol Immunol 1994;9:202–208.
124.
Shinada K, Tagashira M, Watanabe H, Sopapornamorn P, Kanayama A, Kanda T, Ikeda M, Kawaguchi Y: Hop bract polyphenols reduced three-day dental plaque regrowth. J Dent Res 2007;86:848–851.
125.
Shouji N, Takada K, Fukushima K, Hirasawa M: Anticaries effect of a component from shiitake (an edible mushroom). Caries Res 2000;34:94–98.
126.
Silva BB, Rosalen PL, Cury JA, Ikegaki M, Souza VC, Esteves A, Alencar SM: Chemical composition and botanical origin of red propolis, a new type of Brazilian propolis. Evid Based Complement Alternat Med 2008;5:313–316.
127.
Singer AC, Crowley DE, Thompson IP: Secondary plant metabolites in phytoremediation and biotransformation. Trends Biotechnol 2003;21:123–130.
128.
Song JH, Yang TC, Chang KW, Han SK, Yi HK, Jeon JG: In vitro effects of a fraction separated from Polygonum cuspidatum root on the viability, in suspension and biofilms, and biofilm formation of mutans streptococci. J Ethnopharmacol 2007;112:419–425.
129.
Spatafora G, Rohrer K, Barnard D, Michalek S: A Streptococcus mutans mutant that synthesizes elevated levels of intracellular polysaccharide is hypercariogenic in vivo. Infect Immun 1995;63:2556–2563.
130.
Steinberg D, Feldman M, Ofek I, Weiss EI: Effect of a high-molecular-weight component of cranberry on constituents of dental biofilm. J Antimicrob Chemother 2004;54:86–89.
131.
Stookey GK, McDonald JL Jr: Further studies of the cariostatic properties of tin (II) and oat hulls in the rat. J Dent Res 1974;53:1398–1403.
132.
Tagashira M, Uchiyama K, Yoshimura T, Shirota M, Uemitsu N: Inhibition by hop bract polyphenols of cellular adherence and water-insoluble glucan synthesis of mutans streptococci. Biosci Biotechnol Biochem 1997;61:332–335.
133.
Takarada K, Kimizuka R, Takahashi N, Honma K, Okuda K, Kato T: A comparison of the antibacterial efficacies of essential oils against oral pathogens. Oral Microbiol Immunol 2004;19:61–64.
134.
Tamesada M, Kawabata S, Fujiwara T, Hamada S: Synergistic effects of streptococcal glucosyltransferases on adhesive biofilm formation. J Dent Res 2004;83:874–879.
135.
Tang G, Yip HK, Cutress TW, Samaranayake LP: Artificial mouth model systems and their contribution to caries research: a review. J Dent 2003;31:161–171.
136.
Tanzer JM, Freedman ML, Fitzgerald RJ: Virulence of mutants defective in glucosyltransferase, dextran-mediated aggregation, or dextranase activity; in Mergenhagen SE, Rosan B (eds): Molecular Basis of Oral Microbial Adhesion. Washington, American Society for Microbiology, 1985, pp 204–211.
137.
Tanzer JM, Freedman ML, Woodiel FN, Eifert RL, Rinehimer LA: Association of Streptococcus mutans virulence with synthesis of intracellular polysaccharide; in Stiles HM, Loesche WJ, O’Brien TL (eds): Proceedings in Microbiology. Aspects of Dental Caries. London, Information Retrieval, 1976, vol 3, pp 596–616.
138.
Tarsi R, Muzzarelli RA, Guzmán CA, Pruzzo C: Inhibition of Streptococcus mutans adsorption to hydroxyapatite by low-molecular-weight chitosans. J Dent Res 1997;76:665–672.
139.
Touyz LZ, Amsel R: Anticariogenic effects of black tea (Camellia sinensis) in caries-prone rats. Quintessence Int 2001;32:647–650.
140.
Twetman S: Antimicrobials in future caries control? A review with special reference to chlorhexidine treatment. Caries Res 2004;38:223–229.
141.
Vacca-Smith AM, Bowen WH: Binding properties of streptococcal glucosyltransferases for hydroxyapatite, saliva-coated hydroxyapatite, and bacterial surfaces. Arch Oral Biol 1998;43:103–110.
142.
van Loveren C: Sugar alcohols: what is the evidence for caries-preventive and caries-therapeutic effects? Caries Res 2004;38:286–293.
143.
Venkitaraman AR, Vacca-Smith AM, Kopec LK, Bowen WH: Characterization of glucosyltransferase B, GtfC, and GtfD in solution and on the surface of hydroxyapatite. J Dent Res 1995;74:1695–1701.
144.
Verpoorte R, Choi YH, Kim HK: Ethnopharmacology and systems biology: a perfect holistic match. J Ethnopharmacol 2005;100:53–56.
145.
Vogel JJ, Thompson DJ, Phillips PH: Studies on the anticariogenic activity of oat hulls. J Dent Res 1962;41:707–712.
146.
Wen D, Liu Y, Li W, Liu H: Separation methods for antibacterial and antirheumatism agents in plant medicines. J Chromatogr B Analyt Technol Biomed Life Sci 2004;812:101–117.
147.
Wolinsky LE, Mania S, Nachnani S, Ling S: The inhibiting effect of aqueous Azadirachta indica (neem) extract upon bacterial properties influencing in vitro plaque formation. J Dent Res 1996;75:816–822.
148.
Wolinsky LE, Sote EO: Isolation of natural plaque-inhibiting substances from ‘Nigerian chewing sticks’. Caries Res 1984;18:216–225.
149.
Wong L, Sissons C: A comparison of human dental plaque microcosm biofilms grown in an undefined medium and a chemically defined artificial saliva. Arch Oral Biol 2001;46:477–486.
150.
Wu KM, Ghantous H, Birnkrant DB: Current regulatory toxicology perspectives on the development of herbal medicines to prescription drug products in the United States. Food Chem Toxicol 2008;6:2606–2610.
151.
Wunder D, Bowen WH: Action of agents on glucosyltransferases from Streptococcus mutans in solution and adsorbed to experimental pellicle. Arch Oral Biol 1999;44:203–214.
152.
Wu-Yuan CD, Chen CY, Wu RT: Gallotannins inhibit growth, water-insoluble glucan synthesis, and aggregation of mutans streptococci. J Dent Res 1988;67:51–55.
153.
Xiao J, Koo H: Structural organization and dynamics of exopolysaccharide matrix and microcolonies formation by Streptococcus mutans in biofilms. J Appl Microbiol 2010;108:2103–2113.
154.
Xie Q, Li J, Zhou X: Anticaries effect of compounds extracted from Galla chinensis in a multispecies biofilm model. Oral Microbiol Immunol 2008;23:459–465.
155.
Yamanaka A, Kimizuka R, Kato T, Okuda K: Inhibitory effects of cranberry juice on attachment of oral streptococci and biofilm formation. Oral Microbiol Immunol 2004;19:150–154.
156.
Yamashita Y, Bowen WH, Burne RA, Kuramitsu HK: Role of the Streptococcus mutans gtf genes in caries induction in the specific-pathogen-free rat model. Infect Immun 1993;61:3811–3817.
157.
Yanagida A, Kanda T, Tanabe M, Matsudaira F, Oliveira Cordeiro JG: Inhibitory effects of apple polyphenols and related compounds on cariogenic factors of mutans streptococci. J Agric Food Chem 2000;48:5666–5671.
158.
Zero DT: In situ caries models. Adv Dent Res 1995;9:214–230.

J.G.J., P.L.R. and H.K. are members of the Natural Product Research in Oral Biology Group (NatPROB), http://natprob.blogspot.com.

1

This step may not always be included in research studies using natural products since previously characterized bioactive substances may be readily available for testing.

Open Access License / Drug Dosage / Disclaimer
Open Access License: This is an Open Access article licensed under the terms of the Creative Commons Attribution-NonCommercial 3.0 Unported license (CC BY-NC) (www.karger.com/OA-license), applicable to the online version of the article only. Distribution permitted for non-commercial purposes only.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.