ArticlePDF Available

Raffinose Inhibits Streptococcus mutans Biofilm Formation by Targeting Glucosyltransferase

Authors:

Abstract and Figures

Streptococcus mutans is a representative biofilm-forming bacterium that causes dental caries through glucosyltransferase (GTF) activity. Glucans are synthesized from sucrose by GTFs and provide binding sites for S. mutans to adhere tightly to the tooth enamel. Therefore, if a novel compound that interferes with GTF function is developed, biofilm formation control in S. mutans would be possible. We discovered that raffinose, an oligosaccharide from natural products, strongly inhibited biofilm formation, GTF-related gene expression, and glucan production. Furthermore, biofilm inhibition on saliva-coated hydroxyapatite discs through the reduction of bacterial adhesion indicated the applicability of raffinose in oral health. These effects of raffinose appear to be due to its ability to modulate GTF activity in S. mutans. Hence, raffinose may be considered an antibiofilm agent for use as a substance for oral supplies and dental materials to prevent dental caries. IMPORTANCE Dental caries is the most prevalent infectious disease and is expensive to manage. Dental biofilms can be eliminated via mechanical treatment or inhibited using antibiotics. However, bacteria that are not entirely removed or are resistant to antibiotics can still form biofilms. In this study, we found that raffinose inhibited biofilm formation by S. mutans, a causative agent of dental caries, possibly through binding to GtfC. Our findings support the notion that biofilm inhibition by raffinose can be exerted by interference with GTF function, compensating for the shortcomings of existing commercialized antibiofilm methods. Furthermore, raffinose is an ingredient derived from natural products and can be safely utilized in humans; it has no smell and tastes sweet. Therefore, raffinose, which can control S. mutans biofilm formation, has been suggested as a substance for oral supplies and dental materials to prevent dental caries.
Content may be subject to copyright.
Rafnose Inhibits Streptococcus mutans Biolm Formation by
Targeting Glucosyltransferase
So-Young Ham,
a
Han-Shin Kim,
b
Eunji Cha,
a
Taehyeung Lim,
c
Youngjoo Byun,
c
,
d
Hee-Deung Park
a
,
e
a
School of Civil, Environmental, and Architectural Engineering, Korea University, Seoul, South Korea
b
Division of Biotechnology, College of Environmental and Bioresource Sciences, Jeonbuk National University, Iksan, Jeonbuk, South Korea
c
College of Pharmacy, Korea University, Sejong, South Korea
d
Biomedical Research Center, Korea University Guro Hospital, Seoul, South Korea
e
KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, South Korea
ABSTRACT Streptococcus mutans is a representative biolm-forming bacterium that
causes dental caries through glucosyltransferase (GTF) activity. Glucans are synthe-
sized from sucrose by GTFs and provide binding sites for S. mutans to adhere tightly
to the tooth enamel. Therefore, if a novel compound that interferes with GTF func-
tion is developed, biolm formation control in S. mutans would be possible. We dis-
covered that rafnose, an oligosaccharide from natural products, strongly inhibited
biolm formation, GTF-related gene expression, and glucan production. Furthermore,
biolm inhibition on saliva-coated hydroxyapatite discs through the reduction of
bacterial adhesion indicated the applicability of rafnose in oral health. These effects
of rafnose appear to be due to its ability to modulate GTF activity in S. mutans.
Hence, rafnose may be considered an antibiolm agent for use as a substance for
oral supplies and dental materials to prevent dental caries.
IMPORTANCE Dental caries is the most prevalent infectious disease and is expensive
to manage. Dental biolms can be eliminated via mechanical treatment or inhibited
using antibiotics. However, bacteria that are not entirely removed or are resistant to
antibiotics can still form biolms. In this study, we found that rafnose inhibited bio-
lm formation by S. mutans, a causative agent of dental caries, possibly through
binding to GtfC. Our ndings support the notion that biolm inhibition by rafnose
can be exerted by interference with GTF function, compensating for the shortcom-
ings of existing commercialized antibiolm methods. Furthermore, rafnose is an in-
gredient derived from natural products and can be safely utilized in humans; it has
no smell and tastes sweet. Therefore, rafnose, which can control S. mutans biolm
formation, has been suggested as a substance for oral supplies and dental materials
to prevent dental caries.
KEYWORDS rafnose, Streptococcus mutans, biolm, glucosyltransferase
In oral microbial communities, benecial bacteria are advantageous for oral health,
whereas harmful bacteria, which cause various oral diseases, also exist (1). Among
oral diseases, dental caries is the most prevalent infectious disease and is expensive to
manage (2). Left untreated, cavities can lead to inammation of the tissue around the
teeth, infection or abscess formation, and even loss of teeth (3). Bacteria that cause
dental caries are not usually sufciently concentrated to cause problems. However,
when sugar concentrations increase, acidogenic bacteria in oral microbial communities
metabolize sugars to organic acids and synthesize polysaccharides (4). Acidic condi-
tions skew the bacterial community toward dominance by aciduric bacteria, and ele-
vated polysaccharide levels promote biochemical and structural changes in the biolm
matrix (dental plaque) (5, 6).
Editor Justin R. Kaspar, Ohio State University
Copyright © 2022 Ham et al. This is an open-
access article distributed under the terms of
the Creative Commons Attribution 4.0
International license.
Address correspondence to Hee-Deung Park,
heedeung@korea.ac.kr.
The authors declare no conict of interest.
Received 28 October 2021
Accepted 13 April 2022
Published 16 May 2022
May/June 2022 Volume 10 Issue 3 10.1128/spectrum.02076-21 1
RESEARCH ARTICLE
Streptococcus mutans is more viable under acidic conditions than other Streptococcus
species, colonizing the human oral cavity environment (7). Streptococcus mutans is a well-
known causative bacterium of dental caries and utilizes sugars (sucrose, starch, glucose,
and fructose) as sources of carbon (8). Streptococcus mutans secretes glucosyltransferase
(GTF) and fructosyltransferase (FTF) (9). These enzymes synthesize glucan and fructan,
respectively, using sucrose as a substrate, which damage the tooth enamel due to acid pro-
ductionandformationofbiolms (10).
Streptococcus mutans produces three genetically distinct GTFs (GtfB, GtfC, and GtfD).
Each GTF has a unique role but is eventually involved in the formation of dental plaques
(11). GtfB and GtfC are associated with initial microbial adherence and the structural stabil-
ity of the extracellular matrix (12). GtfB synthesizes insoluble glucans rich in
a
-1,3-glycosidic
linkages and is absorbed onto the bacterial cell surface to promote tight cell clustering for
microcolony formation. GtfC, which is adsorbed to the enamel within the pellicle, produces
a mixture of insoluble and soluble glucans with
a
-1,6-linkages and serves as an attachment
site for bacteria. The role of GtfD in plaque formation remains unclear; however, it synthe-
sizes soluble glucans, which serve as primers for GtfB activity (6, 13, 14).
Overall, GTFs synthesize soluble and insoluble glucans from sucrose, permitting the
bacteria to adhere to glucan and subsequently colonize the dental surfaces (12). GTF
enzymes are involved in the formation of glucan, a glucose multimer, accounting for
10 to 20% of dental biolm dry weight (7). Glucan plays an important role in glucan-
cell-tooth surface binding by increasing cell aggregation (11). Streptococcus mutans
can easily adhere to tooth surfaces and contribute to the formation of the biolm and
its structural integrity (15).
To prevent oral diseases such as dental caries, dental biolms are often eliminated
via nonspecic mechanical removal treatments, such as brushing and ossing, or inhib-
ited by inactivating the growth of cariogenic bacteria through the use of antibiotics
(16). Fluoride is the most effective anticaries agent (17); low concentrations of uoride
inactivate a variety of enzymes in intact cells, whereas high concentrations of uoride
enhance the proton permeability of cell membranes as a transmembrane proton car-
rier. Based on these mechanisms, uoride affects the production and tolerance of acid
and the antimicrobial abilities of S. mutans (18). However, continuous and high concen-
trations of uoride treatment cause the emergence of resistant bacteria and uorosis.
Oral bacteria adapted to uoride treatment exhibit stable resistance to high uoride
levels (19). Furthermore, excess uoride treatment hypomineralizes tooth enamel by
increasing the porosity of the surface and subsurface of tooth enamel (20). This alters
the appearance of the tooth, from ne white lines to pitting or staining of enamel.
Therefore, other biolm-inhibiting agents need to be developed to compensate for
these drawbacks.
Recently, some researchers have focused on developing biolm inhibitors that can
interfere with the function of GTFs in S. mutans. Many natural compounds (e.g., hops,
green tea, traditional medicinal plants, and food extracts) have been used as antibio-
lm agents targeting GTFs (2123). Koo et al. found that propolis collected by Apis mel-
lifera bees is a potent inhibitor of GTF enzymes, with high inhibitory activities against
GtfB and GtfC (24). They also discovered that apigenin decreases the accumulation of
S. mutans biolms by affecting the formation of insoluble and soluble glucans in the
polysaccharide matrix (25).
We previously reported that rafnose can reduce Pseudomonas aeruginosa biolm
formation by decreasing cellular cyclic diguanylate levels (26). Rafnose is widely pres-
ent in various vegetables (e.g., ginger, garlic, kohlrabi, onion, parsnip, and scallion) and
fruits (e.g., apricot and melon) (27). It is a trisaccharide consisting of galactose, glucose,
and fructose. Galactose in rafnose can bind to the LecA protein of P. aeruginosa,
which indicates that galactose and rafnose compete for the same binding site of the
LecA protein. Similarly, S. mutans contributes to biolm formation by binding sucrose-
derived glucan to GTFs (11). Therefore, the development of novel compounds that
Inhibition of S. mutans Biolm Formation by Rafnose Microbiology Spectrum
May/June 2022 Volume 10 Issue 3 10.1128/spectrum.02076-21 2
interfere with the binding of glucan to GTFs should be considered to inhibit S. mutans
biolm formation.
The objective of this study was to develop a novel S. mutans biolm inhibitor; we
hypothesized that rafnose affects S. mutans biolm formation by reducing the activity
of GTFs. We investigated S. mutans biolm formation and the molecular mechanisms
with rafnose treatment to prove the hypothesis. Biolm formation was tested under
both static and ow conditions, and the antibiolm mechanism was deduced by esti-
mating sucrose consumption and GtfC binding afnity. GTF-related gene expression
levels and glucan production were measured using reverse transcription-quantitative
PCR (RT-qPCR) and colorimetric methods, respectively. Furthermore, the applicability
of rafnose to dental caries was evaluated using bacterial adhesion tests and scanning
electron microscopy (SEM) analysis of biolms formed on articial-saliva-coated hy-
droxyapatite (HA) discs.
RESULTS
Effects of rafnose on S. mutans biolm formation. Rafnose reduced biolm for-
mation by most Streptococcus species (KCOM 1136 to KCOM 1228) isolated from human
mouths (Fig. 1). Biolm formation by S. mutans and S. sobrinus was inhibited, on average,
by 12 to 25% and 11 to 17%, respectively, compared to untreated biolm (i.e., control
biolm), when treated with 100 or 1,000
m
Mrafnose. In particular, biolm formation by
S. mutans KCOM 1136, a representative biolm-forming bacterium in this study, was
decreased most signicantly following rafnose treatment, which resulted in biolm forma-
tion being reduced by .50%, compared to control biolm, after 1,000
m
Mrafnose
treatment.
The inhibition of KCOM 1136 biolm formation by rafnose treatment was analyzed
under static and ow conditions. Under static conditions, biolm formation was dra-
matically decreased by 44% at a high concentration of rafnose (1,000
m
M) (Fig. 2A).
Under ow conditions, although the morphology of the biolm seemed to be similar,
with a bumpy shape in control and rafnose-treated biolms, the average volume and
thickness of the rafnose-treated biolm were decreased by 54 to 64%, compared to
the control biolm (Fig. 2B).
Rafnose is a trisaccharide consisting of galactose and sucrose (see Fig. S1A in the sup-
plemental material). However, contradictory patterns in S. mutans biolm formation were
observed for sucrose and galactose treatments. Galactose exhibited biolm-inhibiting activ-
ity similar to that of rafnose; in contrast, biolm formation was enhanced in the presence
of sucrose (see Fig. S1B). Moreover, sucrose increased S. mutans growth at a concentration
of 1,000
m
M (see Fig. S2), while there were no differences in the growth of S. mutans with
rafnose treatment (0 to 1,000
m
M).
Mechanism of S. mutans biolm inhibition by rafnose. Sucrose consumption of
S. mutans following rafnose treatment was monitored for 24 h to investigate the
FIG 1 Biolm formation of Streptococcus species following rafnose treatment for 24 h. Biolms of S.
mutans and S. sobrinus were formed following rafnose treatment (0 to 1,000
m
M) under static
conditions. Error bars indicate the standard deviations of ve measurements. **,P,0.005; *,
P,0.05, versus the control. Raf, rafnose.
Inhibition of S. mutans Biolm Formation by Rafnose Microbiology Spectrum
May/June 2022 Volume 10 Issue 3 10.1128/spectrum.02076-21 3
relationship between rafnose and sucrose. The method used for assessment of sucrose
consumption was suitable for detecting sucrose concentration but did not respond to
high concentrations of rafnose, as indicated in the standard curves for sucrose and raf-
nose (see Fig. S3). Streptococcus mutans consumed 88% of the sucrose over 24 h
(Fig. 3A). However, as the concentrations of rafnose with sucrose in S. mutans increased,
sucrose consumption decreased, compared to that in the control (i.e., no rafnose treat-
ment). When 10, 100, and 1,000
m
Mrafnose was used together with 100
m
Msucrose,
79%, 70%, and 56% of the sucrose, respectively, was consumed over 24 h.
Furthermore, competitive biolm formation was analyzed by adding various con-
centrations of rafnose and sucrose (0 to 1,000
m
M) simultaneously to the culture me-
dium of S. mutans. Biolm formation decreased by 50% following treatment with
1,000
m
M rafnose, whereas it decreased by 26% with simultaneous treatment with
FIG 2 Streptococcus mutans biolm formation following rafnose treatment under static and ow conditions. (A) CV-
stained biolm following rafnose treatment (0 to 1,000
m
M). Quantication was performed by measuring the OD under
static conditions (OD
545
/OD
595
). Error bars indicate the standard deviations of ve measurements. **,P,0.005, versus the
control. Raf, rafnose. (B) Volume and thickness of DAPI-stained biolm based on CLSM images; 1,000
m
M rafnose was
added to the S. mutans biolm for 48 h under ow conditions. Raf, Rafnose.
Inhibition of S. mutans Biolm Formation by Rafnose Microbiology Spectrum
May/June 2022 Volume 10 Issue 3 10.1128/spectrum.02076-21 4
FIG 3 Possible mechanisms underlying the inhibition of Streptococcus mutans biolm formation following rafnose
treatment. (A) Sucrose consumption in S. mutans biolm cells treated with rafnose. Streptococcus mutans biolm was
formed following sucrose (100
m
M) and rafnose (0 to 1,000
m
M) treatments under static conditions for 24 h. Error bars
indicate the standard deviations of three measurements. **,P,0.005, versus the control. Raf, rafnose; Suc, sucrose. (B)
Competitive biolm formation tests between rafnose and sucrose. Streptococcus mutans biolm was formed following
treatment with rafnose and sucrose at concentrations of 0 to 1,000
m
M. Error bars indicate the standard deviations of
ve measurements. **,P,0.005; *,P,0.05, versus the control. (C) Best-docked poses of rafnose in S. mutans
glucansucrase (GtfC [PDB code 3AIC]).
Inhibition of S. mutans Biolm Formation by Rafnose Microbiology Spectrum
May/June 2022 Volume 10 Issue 3 10.1128/spectrum.02076-21 5
1,000
m
M sucrose, compared to that in the control (i.e., no rafnose treatment) (Fig. 3B,
left). This nding indicated that the inhibition of biolm formation following rafnose
treatment decreased as the sucrose concentration increased. Similarly, as the rafnose
concentration increased, biolm formation following sucrose treatment decreased
(Fig. 3B, right). These results suggest that rafnose and sucrose have a competitive
relationship regarding sucrose consumption and biolm formation in S. mutans.
The possibility of rafnose acting as a glucansucrase inhibitor in S. mutans was investi-
gated using molecular docking studies. Figure 3C shows the best-docked poses of rafnose
intheactivesiteoftheS. mutans glucansucrase.Thedockedposeofrafnose was tted
inside the active site of glucansucrase and formed hydrogen bonds with Asp593, a key
amino acid residue considered to be the most critical point for acceptor sugar orientation,
which inuences the transglycosylation specicity of S. mutans glucansucrase (28).
However, the docked pose of sucrose was located only in subunit 1, which consists of Arg
475, Asp477, Glu515, and Asp909, without interaction with Asp593, as shown in Fig. S4A in
the supplemental material. The glucosyl and fructosyl moieties, which are common in both
rafnose and sucrose, made close contact with the amino acid residues of the subunit 1
site. For D-galactose, the best-docked pose was also in close contact only with the amino
acid residues of subunit 1, including Asp475, Asp477, Asp909, and Gln 960 (see Fig. S4B). In
particular, the OH groups at the C-3 and C-4 positions of the galactose moiety in rafnose
interacted with Asp593 as well as with the amino acid residues of the subunit 1 site, which
might enhance its binding afnity for glucansucrase. Overall, the docking studies sug-
gested that rafnose, sucrose, and galactose might be in contact with the subunit 1 site of
glucansucrase, with total scores of 9.90, 7.88, and 6.42, respectively.
Effects of rafnose on GTFs in S. mutans.GTF-related gene expression levels and
glucan production were analyzed to assess the effects of rafnose on GTFs in S.
mutans. As shown in Fig. 4A, all GTF-related genes were signicantly downregulated in
S. mutans biolm cells treated with 1,000
m
M rafnose. The expression of gtfB,gtfC,
and gtfD was repressed by 69%, 74%, and 38%, respectively, compared to that in the
control (S. mutans biolm cells without rafnose treatment). Galactose also evenly
decreased GTF-related gene expression by 40 to 43%, whereas sucrose increased the
expression levels of GTF-related genes, especially gtfC and gtfD, by 1.9- to 5.5-fold,
compared to those in the control (see Fig. S5A and B). Furthermore, when rafnose
and sucrose were used together, most GTF-related gene levels were not signicantly
different from those in the control (see Fig. S5C). The expression of the 16S rRNA refer-
ence gene was not signicantly affected by any of the treatments.
FIG 4 GTF-related changes in Streptococcus mutans following rafnose treatment. (A) GTF gene expression levels in S. mutans biolm cells. Streptococcus
mutans biolm was formed following rafnose treatment (1,000
m
M) under static conditions for 24 h. Relative fold changes were evaluated by RT-qPCR
analysis. Error bars indicate the standard deviations of ve measurements. **,P,0.005; *,P,0.05, versus the control. Raf, rafnose. (B) Relative glucan
production of S. mutans following rafnose treatment. Extracted insoluble glucan was reacted with rafnose (0 to 1,000
m
M). Glucan production was
evaluated using the colorimetric method. Error bars indicate the standard deviations of three measurements. **,P,0.005, versus the control. Raf, rafnose.
Inhibition of S. mutans Biolm Formation by Rafnose Microbiology Spectrum
May/June 2022 Volume 10 Issue 3 10.1128/spectrum.02076-21 6
To evaluate the production of water-insoluble glucans, GTFs were extracted, using
solid ammonium sulfate, from S. mutans cultured in brain heart infusion (BHI) medium
containing sucrose. The precipitated GTFs were then reacted with rafnose (0 to
1,000
m
M). Using sucrose as a substrate, the production of glucans was estimated by
measuring the intensity of glucan color using a spectrophotometer. There were no sig-
nicant changes in glucan production in S. mutans at concentrations below 10
m
M raf-
nose (Fig. 4B). However, glucan production was reduced by 18 to 39% after rafnose
treatment at 100 to 1,000
m
M, in a concentration-dependent manner. These results
implied that rafnose affected the downregulation of GTF-related gene expression and
decreased glucan production in S. mutans (Fig. 5).
Application of rafnose on articial-saliva-coated HA discs. Articial-saliva-coated
HA discs were used to simulate conditions of the oral environment in humans.
Adhesion of S. mutans was evaluated by staining and counting biolm cells on HA
discs. As shown in Fig. 6A, S. mutans biolm cells on HA discs treated with 1,000
m
M
rafnose were not stained well, compared to the control, which indicated that S.
mutans cells had difculties in adhering to HA discs after rafnose treatment. Similar
results were observed with the colony-counting method, in which the number of colo-
nies on HA discs was diminished following rafnose treatment (Fig. 6B).
The effects of rafnose on S. mutans biolm formation on HA discs were also dem-
onstrated via SEM analysis. There was no difference between HA discs coated with or
without articial saliva in the SEM images (Fig. 7A and B). Figure 7C and D show SEM
images of S. mutans biolms formed on HA discs. Biolm formation after 1,000
m
M raf-
nose treatment was reduced signicantly, compared to the control (no rafnose treat-
ment). Galactose also adversely affected biolm formation, whereas sucrose increased
it (see Fig. S6). This nding indicates that rafnose can effectively control S. mutans
FIG 5 GTF-mediated biolm inhibition in Streptococcus mutans following rafnose treatment. GTFs
secreted by S. mutans are adsorbed on the pellicle and bacterial surfaces. The adsorbed GTFs bind to
sucrose-derived glucan. Glucan provides binding sites on the surfaces for S. mutans, mediating
adherence to the tooth enamel and tight bacterial clustering and eventually promoting biolm
formation. However, if the S. mutans biolm is treated with rafnose, then the rafnose is expected
to prevent sucrose-derived glucan from binding to GTFs. Therefore, the activity of glucan production
in S. mutans is reduced, which may retard biolm formation.
Inhibition of S. mutans Biolm Formation by Rafnose Microbiology Spectrum
May/June 2022 Volume 10 Issue 3 10.1128/spectrum.02076-21 7
biolm formation under conditions similar to those of the human oral environment.
However, these results do not reliably indicate the applicability of rafnose in the treat-
ment of dental caries. To prevent or treat dental caries with rafnose, further research
should be conducted, including clinical demonstration and development of efcient
methods for rafnose application.
DISCUSSION
In this study, we discovered that rafnose reduced biolm formation by bacteria that
cause dental caries in humans. The common cariogenic bacteria, S. mutans and S. sobrinus,
adhere to the enamel salivary pellicle and other plaque bacteria to form biolms (29).
Rafnose inhibited S. mutans biolm formation more effectively than S. sobrinus biolm for-
mation (Fig. 1). S. mutans is predominantly found in dental biolm development (74 to
94% of the diverse carious bacterial population), whereas S. sobrinus is less prevalent and is
FIG 6 Adhesion of Streptococcus mutans to articial-saliva-coated HA discs following rafnose treatment
(1,000
m
M). (A) Bacterial adhesion evaluation using CV staining. Streptococcus mutans biolm was formed on
HA discs for 24 h under static conditions, and OD
545
was assessed. Error bars indicate the standard deviations
of three measurements. *,P,0.05, versus the control. Raf, rafnose. (B) Bacterial adhesion evaluation using
the cell-counting method. The number of colonies of separated biolm cells that formed on the HA discs was
calculated using the standard plate culture method. Error bars indicate the standard deviations of three
measurements. **,P,0.005, versus the control.
FIG 7 SEM images of Streptococcus mutans biolm cells following rafnose treatment (1,000
m
M) on
articial-saliva-coated HA discs. (A) HA discs. (B) Articial-saliva-coated HA discs. (C) Streptococcus
mutans biolm cells on articial-saliva-coated HA discs. (D) Rafnose-treated S. mutans biolm cells
on articial-saliva-coated HA discs.
Inhibition of S. mutans Biolm Formation by Rafnose Microbiology Spectrum
May/June 2022 Volume 10 Issue 3 10.1128/spectrum.02076-21 8
usually detected along with S. mutans (30). S. mutans mainly synthesizes polysaccharides
from sucrose as a substrate, contributing to an increase in bacterial cell surface hydropho-
bicity and biolm formation (31). Streptococcus mutans biolm formation increased in
response to sucrose, as shown in Fig. S1B in the supplemental material.
Sucrose accelerates S. mutans biolm formation by targeting GTFs (32). Our RT-qPCR
results showed that rafnose mainly downregulated the expression of gtfB and gtfC in
S. mutans (Fig.4A).However,gtfD expression slightly decreased after rafnose treatment.
GtfB and GtfC are highly homologous and share 75% amino acid sequence identity in an
operon-like arrangement. In contrast, GtfD, which is not linked to the gtfBC locus, shares
,50% identity with GtfB and GtfC (25). The different patterns for gtfB-C and gtfD may be
related to various factors (carbohydrate availability and source, environmental pH, and
growth rate/phase) that inuence the transcription of gtf genes (25).
Streptococcus mutans synthesizes sticky, water-insoluble/soluble extracellular glu-
cans from sucrose as the sole substrate via the production of GTFs (33). Sticky glucans
enhance the pathogenic potential of dental plaque by providing binding sites for
S. mutans colonization and promoting accumulation on tooth surfaces. A large propor-
tion of glucans contribute to the establishment of the extracellular polysaccharide
matrix and provide bulk and structural integrity to dental biolms (24, 25). In particular,
insoluble glucans are directly related to bacterial attachment during biolm formation
by S. mutans in dental caries. In contrast, soluble glucans indirectly contribute to bio-
lm formation as a nutrient source for biolm cells (31). The production of insoluble
glucans decreased in proportion to the concentration of rafnose (Fig. 4B).
Most studies regarding biolm inhibitors for dental caries have focused largely on
enzymatic activity in solution, without considering the importance of enzymes
adsorbed on the pellicle of the tooth surface. Current commercially available antibio-
lm agents show high levels of resistance to surface-adsorbed GTF enzymes; thus, it
should be considered that inhibitors of GTF function effectively under conditions simi-
lar to those of the oral environment (34). In this study, we used saliva-coated HA, the
main constituent of tooth enamel, because GTFs are detected in whole saliva at high
levels in individuals with dental caries (11). Furthermore, saliva-coated HA can be
tightly bound with GTFs, although GTFs have a low binding afnity for HA and lose
their activity over time. As shown in Fig. 7, S. mutans biolm formation on saliva-coated
HA discs was decreased following rafnose treatment.
Although all GTFs interact with saliva-coated HA discs, GtfC has a higher binding af-
nity, compared with those of GtfB and GtfD (12). GtfC is signicantly adsorbed onto
the pellicle when exposed to sucrose, thereby enhancing the adherence of bacterial
cells to dental surfaces (35). Therefore, inhibition of GTF activities, especially those of
GtfC and mainly those adsorbed to the salivary pellicle, is prioritized to prevent the for-
mation of pathogenic dental plaque (24). GtfC secreted by S. mutans is commonly
incorporated into the pellicle, and surface-adsorbed GtfC utilizes sucrose to produce
glucans (11). Glucans provide binding sites on the surface of S. mutans to easily adhere
to tooth enamel, facilitating biolm formation. In our study, rafnose treatment
degraded glucan production, and S. mutans had difculty adhering to saliva-coated HA
discs, eventually resulting in inhibition of biolm formation on HA discs (Fig. 5).
This phenomenon might be related to the interruption of GTF by rafnose. Streptococcus
mutans consumes sucrose to synthesize glucan, thereby increasing biolm formation (11).
When rafnose was combined with sucrose in S. mutans, sucrose consumption and biolm
formation decreased (Fig. 3A), suggesting that rafnose may control GTF activity in S.
mutans. In particular, this may be related to the possibility of rafnose binding to GtfC
(Fig. 3C). Galactose in rafnose has additional interactions with Asp593, a key amino acid res-
idue, and amino acid residues of the subunit 1 site, which might enhance its binding afnity
for glucansucrase. Furthermore, galactose showed inhibitory activities similar to those of raf-
nose in biolm formation and GTF gene expression in S. mutans, whereas sucrose showed
the opposite effects (Fig. S1B and Fig. S5). Ryu et al. also discovered that galactose signi-
cantly inhibits S. mutans biolm formation by decreasing the expression of three GTF genes
Inhibition of S. mutans Biolm Formation by Rafnose Microbiology Spectrum
May/June 2022 Volume 10 Issue 3 10.1128/spectrum.02076-21 9
(36). These results suggest that galactose may play an important role in the various activities
of rafnose in S. mutans. However, these results do not provide direct evidence that raf-
nose binds to GtfC. Further studies are required to elucidate the detailed mechanism by
which rafnose affects GTF activity in S. mutans.
Nagasawa et al. reported that rafnose induces S. mutans biolm formation with
low concentrations of sucrose, which is contrary to our ndings (31). Although there
may be many reasons for the difference in biolm formation between the two studies,
we cautiously suggest that the difference is correlated with the concentration of raf-
nose. Streptococcus mutans biolm formation decreased in proportion to the rafnose
concentration (0 to 1,000
m
M) in our study. However, the inhibition of S. mutans bio-
lm formation by rafnose treatment decreased as the sucrose concentration
increased (see Fig. S7A). This can be explained by the fact that rafnose is insufcient
to inhibit biolm formation at high levels of sucrose but this does not decrease the
activity of rafnose. Conversely, rafnose treatment above 0.03% increases S. mutans
biolm formation (31). A rafnose concentration of 0.03% is equal to 620
m
M, which is
relatively higher than the rafnose concentrations used in our study. This difference
was expected to be due to the remaining rafnose used to inhibit S. mutans biolm
formation. Rafnose can serve as a substrate for FTF in Streptococcus species but is not
used by GTF (3739). Nagasawa et al. also found that rafnose induces S. mutans bio-
lm formation through fructan synthesis (31). Furthermore, rafnose treatment gener-
ally shows biolm formation patterns; however, at a concentration of less than 0.03%
rafnose, there are slight biolm inhibition patterns. Similarly, in this study, S. mutans
biolm formation increased following treatment with millimolar concentrations of raf-
nose (see Fig. S7B). Therefore, determination of the rafnose concentrations suitable
for environmental conditions should be considered when applying rafnose to regu-
late S. mutans biolm formation.
Rafnose, an oligosaccharide found in natural products, greatly reduced S. mutans bio-
lm formation under static and ow conditions. Furthermore, GTF-related gene expression
levels and glucan production were decreased following 1,000
m
Mrafnose treatment.
Moreover, the reduction in bacterial adhesion following rafnose treatment delayed S.
mutans biolm formation on saliva-coated HA discs. The activities of rafnose can be
explained by the possibility of binding to GtfC by rafnose.Hence,rafnose has the poten-
tial to be utilized as a natural substance to prevent S. mutans biolmformationintheoral
environment.
MATERIALS AND METHODS
Bacteria and chemicals. S. mutans (strain KCOM 1136; Korean Collection for Oral Microbiology
[KCOM]) used in this study was clinically isolated and identied in the oral microbiota of Korean subjects.
Streptococcus mutans cultured overnight was prepared by incubation with BHI medium (Difco, Detroit,
MI, USA) at 37°C in a shaking incubator (200 rpm).
D-(1)-Rafnose pentahydrate, sucrose, and D-(1)-galactose were selected as chemicals that affect S.
mutans biolm formation. These chemicals were purchased from Sigma-Aldrich (St. Louis, MO, USA) and
dissolved in dimethyl sulfoxide (DMSO) (Carl Roth, Karlsruhe, Germany).
Static biolm formation test. Streptococcus mutans cultured overnight was adjusted to an optical
density at 595 nm (OD
595
) of 1.0 using a UV-visible spectrophotometer (UV mini-1240; Shimadzu, Kyoto,
Japan) and then diluted 1:20 (vol/vol) in BHI medium containing 10
m
M sucrose. The diluted bacterial
culture was grown in the presence of 0 to 1,000
m
M sugars for 24 h in 96-well polystyrene microtiter
plates (Sigma-Aldrich) and borosilicate bottles. The plates and bottles were incubated at 37°C without
agitation, to allow biolm formation. After 24 h of incubation, the OD
595
of the suspended cultures in
the plates and bottles was measured using an iMark microplate reader (Bio-Rad, Richmond, CA, USA) to
analyze the growth of the bacterial cells. The suspended culture was then discarded, and biolm cells
attached to the plates and bottles were washed with phosphate-buffered saline (PBS) (137 mM NaCl,
2.7 mM KCl, 10 mM Na
2
HPO
4
, and 2 mM KH
2
PO
4
[pH 7.2]) to remove the remaining suspended culture
medium. Biolm cells were stained with 0.1% crystal violet (CV) for 30 min. After the staining period, CV
was washed using sterile deionized water to remove the remaining dye and dissolved in 100% ethyl
alcohol. The remaining CV was measured at 545 nm using a microplate reader. The extent of CV staining
indicated the amount of biolm formed. Biolm formation was normalized by dividing OD
545
by OD
595
.
Flow biolm formation test. To form biolms under ow conditions, glass slides were inserted into a
drip-ow reactor (DFR-110; BioSurface Technologies Corp., Bozeman, MT, USA). BHI medium containing a
dilution of S. mutans cultured overnight (OD
595
of 1.0) with or without rafnose (1,000
m
M) was fed into the
reactor using a peristaltic pump (Masterex C/L tubing pump; Cole-Parmer, Vernon Hills, IL, USA) at a ow
Inhibition of S. mutans Biolm Formation by Rafnose Microbiology Spectrum
May/June 2022 Volume 10 Issue 3 10.1128/spectrum.02076-21 10
rate of 0.3 mL/min. The reactor was operated at 37°C for 48 h. At the end of the reactor operation, the slides
were carefully removed from the reactor. Biolms formed on the slides were washed with PBS twice and
stained with 49,6-diamidino-2-phenylindole (DAPI) (Carl Roth) for confocal laser scanning microscopy (CLSM)
(LSM 700; Carl Zeiss, Jena, Germany) analysis.
Biolms were measured in z-stack mode under blue uorescent light (excitation wavelength, 350 nm;
emission wavelength, 470 nm) with a 20lens objective (W N-Achroplan 20/0.5 W [DIC] M27). Biolm mor-
phology was analyzed using ZEN 2011 software (Carl Zeiss), and the volume and thickness of biolms were
measured using the Comstat2 tool of ImageJ software (National Institutes of Health, Bethesda, MD, USA) (40).
Sucrose consumption test. A sucrose consumption test was conducted by modifying the method
presented in a previous study to suit the experimental conditions (41). A dilution of S. mutans cultured
overnight (OD
595
of 0.05) in BHI medium treated with sucrose (100
m
M) and rafnose (10 to 1,000
m
M)
was aliquoted in borosilicate bottles at 37°C for 24 h. To calculate sucrose consumption, bacterial sus-
pensions were sampled every 6 h and passed through a 0.22-
m
m membrane lter. Sucrose consumption
by the suspension was examined using a sucrose assay kit (Sigma-Aldrich). The ltered suspension
(100
m
L) was incubated at 25°C for 10 min with 100
m
L of sucrose assay reagent. After the addition of
2,000
m
L of glucose assay reagent, the reaction mixture was incubated again at 25°C for 15 min. Sucrose
consumption was evaluated by measuring the OD
340
using a spectrophotometer.
In silico docking studies. All compounds were rendered as two-dimensional (2D) and three-dimensional
(3D) structures using ChemDraw Ultra v.12.0.2.1076 and Chem3D Pro v.12.0.2, respectively. Ligand preparation
and optimization were performed using the Sanitize preparation protocol in SYBYL-X v.2.1.1 (Tripos Inc., St.
Louis, MO, USA). The GtfC protein structure (Protein Data Bank [PDB] code 3AIC) in PDB format was down-
loaded from the Research Collaboratory for Structural Bioinformatics (RCSB) PDB. The SYBYL-X v.2.1.1 program
was employed for protein preparation, including xation of conicting side chains of amino acid residues.
Water molecules were removed from the protein crystal structure, and chains other than chain A were also
removed. Hydrogen atoms were added under the application of AMBER7 FF09 for the force eld setting. The
minimization process was performed using the POWELL method. The initial optimization option was set to
zero. Docking studies involving the ligands were performed using the Surex-Dock GeomX module in SYBYL-
X v.2.1.1. Docking was guided by the Surex-Dock protocol, and the docking site was dened by the Ligand
method with the complexed ligand
a
-acarbose with a threshold value of 0.50. Other parameters were applied
with default settings in all runs.
RT-qPCR. A dilution of S. mutans cultured overnight (OD
595
of 0.05) in BHI medium containing 10
m
Msu-
crose and treated with or without rafnose (1,000
m
M) was incubated in borosilicate bottles at 37°C for 24 h.
Biolm samples were collected by scraping bacterial cells attached to the bottles with PBS. Total RNA was
extracted from the biolm samples using TRI Reagent (Molecular Research Center, Cincinnati, OH, USA). RT-
qPCR was performed with SYBR Premix Ex Taq (TaKaRa, Shiga, Japan) on a Bio-Rad CFX 96 real-time PCR sys-
tem (Bio-Rad). The primer sets for GTF-related genes were designed using Primer 3 v.0.4.0 (http://frodo.wi.mit
.edu) (see Table S1 in the supplemental material). Thermocycling conditions for the designed primer sets
were as follows: initial denaturation at 95°C for 10 s, followed by 40 cycles of denaturation at 95°C for 10 s,
annealing at 60°C for 10 s, and extension at 63°C for 34 s. Fluorescent signal intensities were measured at the
end of the nal extension step. Relative gene expression was normalized to that of 16S rRNA as a reference
gene (42) and analyzed using the 2
2DDCT
relative expression method (43).
Glucan production test. A glucan production test was conducted by modifying the methods used
in previous studies (44, 45). A dilution of S. mutans cultured overnight (OD
595
of 0.01) in BHI medium
containing 10
m
M sucrose was incubated at 37°C for 24 h with shaking (200 rpm). The supernatant of
incubated S. mutans obtained by centrifugation at 4°C for 30 min was precipitated with solid ammonium
sulfate (Sigma-Aldrich). After agitation at 4°C for 1 h and centrifugation at 10,000 gat 4°C for 30 min,
the precipitate was diluted with 10 mM potassium phosphate buffer (pH 6.0). The extracted crude GTFs
were stored at 280°C.
To measure the effects of rafnose treatment on water-insoluble glucan production, mixtures of GTFs
treated with rafnose (0 to 1,000
m
M) and 20
m
L of 0.0625 M potassium phosphate buffer (pH 6.5) were
reacted with 12.5
m
g/L sucrose and 0.25
m
g/L sodium azide (Sigma-Aldrich) and then incubated at 37°C for
24 h. Precipitated water-insoluble glucans were dispersed using a sonicator (VCX 750; SONICS, Newtown, CT,
USA) for 4 cycles of 5 s of operation and 5 s of pause, at a frequency of 3.5 Hz. The glucan production was
estimated by measuring the OD
550
using a UV-visible spectrophotometer.
Bacterial adhesion test. Dense ceramic HA discs (Clarkson Chromatography Products, Williamsport,
PA, USA) were coated with articial saliva. Following an incubation period at 25°C for 12 h, the HA discs
were rotated at 5 rpm for 1 h with articial saliva. Articial saliva was prepared according to the ISO/
TR10271 standard, i.e., 0.04% sodium chloride (NaCl), 0.08% calcium chloride dihydrate (CaCl
2
2H
2
O),
0.04% potassium chloride (KCl), 0.0005% sodium sulde dihydrate (Na
2
S2H
2
O), 0.08% sodium dihydro-
gen phosphate (NaH
2
PO
4
2H
2
O), and 0.1% urea (CH
4
N
2
O) (30). The coated discs were then washed for
30 min with buffered KCl containing 5 mg/mL bovine serum albumin.
A dilution of S. mutans cultured overnight (OD
595
of 0.01) in BHI medium containing 10
m
M sucrose
was adhered to articial-saliva-coated HA discs by incubating the bacterial cells with or without rafnose
treatment (0 to 1,000
m
M) at 37°C for 1.5 h. Adherent bacterial cells on HA discs were washed twice with
PBS and then analyzed using CV staining and cell-counting methods. CV staining was performed as
described above for the static biolm formation test. For cell counting, HA discs were sonicated for
10 min at an amplitude of 30%, using a sonicator, to detach bacterial cells from the discs. The detached
bacterial cells were dispersed, diluted, spread on BHI agar plates, and then incubated at 37°C for 24 h.
CFU were calculated by counting the number of bacterial colonies formed on agar plates.
Inhibition of S. mutans Biolm Formation by Rafnose Microbiology Spectrum
May/June 2022 Volume 10 Issue 3 10.1128/spectrum.02076-21 11
SEM analysis. A dilution of S. mutans cultured overnight (OD
595
of 0.05) in BHI medium containing
10
m
M sucrose was incubated with glass slides at 37°C for 24 h. Biolm cells on the slides were xed
with 4% glutaraldehyde solution (Sigma-Aldrich) at 4°C for 1 h. Then, the xed biolm cells were sequen-
tially dehydrated in 50%, 80%, and 100% ethyl alcohol for 20 min each. Dehydrated biolm cells were
dried in a vacuum desiccator for 12 h and coated with platinum for 90 s using an E-1030 ion-sputter
coater (Hitachi, Tokyo, Japan). The coated sample was observed using a model S-4300 scanning electron
microscope (Hitachi) operated at a magnication of 5,000 and a voltage of 15.0 kV.
Statistical analysis. Pvalues were estimated via Student's ttest using SigmaPlot (Systat Software,
Inc., San Jose, CA, USA).
SUPPLEMENTAL MATERIAL
Supplemental material is available online only.
SUPPLEMENTAL FILE 1, PDF le, 0.7 MB.
ACKNOWLEDGMENTS
This work was supported by the National Research Foundation of Korea (grant
2020R1A6A1A03045059 to H-D.P. and grant 2019R1A6A1A03031807 to Y.B).
REFERENCES
1. Kolenbrander PE, Andersen RN, Blehert DS, Egland PG, Foster JS, Palmer
RJ, Jr. 2002. Communication among oral bacteria. Microbiol Mol Biol Rev
66:486505. https://doi.org/10.1128/MMBR.66.3.486-505.2002.
2. Marsh PD. 2003. Are dental diseases examples of ecological catastrophes? Mi-
crobiology (Reading) 149:279294. https://doi.org/10.1099/mic.0.26082-0.
3. Laudenbach JM, Simon Z. 2014. Common dental and periodontal diseases:
evaluation and management. Med Clin North Am 98:12391260. https://doi
.org/10.1016/j.mcna.2014.08.002.
4. Scheie AA, Petersen FC. 2004. The biolm concept: consequences for future
prophylaxis of oral diseases? Crit Rev Oral Biol Med 15:412. https://doi.org/
10.1177/154411130401500102.
5. Marsh PD. 2006. Dental plaque as a biolm and a microbial community:
implications for health and disease. BMC Oral Health 6:S14. https://doi
.org/10.1186/1472-6831-6-S1-S14.
6. XiaoJ,KleinMI,FalsettaML,LuB,DelahuntyCM,YatesIIJ,HeydornA,KooH.
2012. The exopolysaccharide matrix modulates the interaction between 3D
architecture and virulence of a mixed-species oral biolm. PLoS Pathog 8:
e1002623. https://doi.org/10.1371/journal.ppat.1002623.
7. Krzy
sciak W, Jurczak A, Ko
scielniak D, Bystrowska B, Skalniak A. 2014. The
virulence of Streptococcus mutans and the ability to form biolms. Eur J
Clin Microbiol Infect Dis 33:499515. https://doi.org/10.1007/s10096-013
-1993-7.
8. Duarte S, Klein M, Aires C, Cury J, Bowen W, Koo H. 2008. Inuences of starch
and sucrose on Streptococcus mutans biolms. Oral Microbiol Immunol 23:
206212. https://doi.org/10.1111/j.1399-302X.2007.00412.x.
9. Hamada S, Koga T, Ooshima T. 1984. Virulence factors of Streptococcus
mutans and dental caries prevention. J Dent Res 63:407411. https://doi
.org/10.1177/00220345840630031001.
10. Duguid R. 1985. In-vitro acid production by the oral bacterium Strepto-
coccus mutans 10449 in various concentrations of glucose, fructose
and sucrose. Arch Oral Biol 30:319324. https://doi.org/10.1016/0003
-9969(85)90004-4.
11. Bowen WH, Koo H. 2011. Biology of Streptococcus mutans-derived gluco-
syltransferases: role in extracellular matrix formation of cariogenic bio-
lms. Caries Res 45:6986. https://doi.org/10.1159/000324598.
12. Ren Z, Chen L, Li J, Li Y. 2016. Inhibition of Streptococcus mutans polysac-
charide synthesis by molecules targeting glycosyltransferase activity. J
Oral Microbiol 8:31095. https://doi.org/10.3402/jom.v8.31095.
13. Aoki H, Shiroza T, Hayakawa M, Sato S, Kuramitsu H. 1986. Cloning of a
Streptococcus mutans glucosyltransferase gene coding for insoluble glu-
can synthesis. Infect Immun 53:587594. https://doi.org/10.1128/iai.53.3
.587-594.1986.
14. Hanada N, Kuramitsu HK. 1988. Isolation and characterization of the Strep-
tococcus mutans gtfC gene, coding for synthesis of both soluble and insol-
uble glucans. Infect Immun 56:19992005. https://doi.org/10.1128/iai.56
.8.1999-2005.1988.
15.YamashitaY,BowenWH,BurneRA,KuramitsuHK.1993.RoleoftheStrepto-
coccus mutans gtf genes in caries induction in the specic-pathogen-free rat
model. Infect Immun 61:38113817. https://doi.org/10.1128/iai.61.9.3811
-3817.1993.
16. Emilson CG. 1994. Potential efcacy of chlorhexidine against mutans
streptococci and human dental caries. J Dent Res 73:682691. https://doi
.org/10.1177/00220345940730031401.
17. Hardwick K, Barmes D, Writer S, Richardson LM. 2000. International collabora-
tive research on uoride. J Dent Res 79:893904. https://doi.org/10.1177/
00220345000790040301.
18. Koo H. 2008. Strategies to enhance the biological effects of uoride on dental
biolms. Adv Dent Res 20:1721. https://doi.org/10.1177/154407370802000105.
19. Liao Y, Brandt BW, Li J, Crielaard W, Van Loveren C, Deng DM. 2017. Fluo-
ride resistance in Streptococcus mutans: a mini review. J Oral Microbiol 9:
1344509. https://doi.org/10.1080/20002297.2017.1344509.
20. Browne D, Whelton H, O'Mullane D. 2005. Fluoride metabolism and uo-
rosis. J Dent 33:177186. https://doi.org/10.1016/j.jdent.2004.10.003.
21. Wu-Yuan CD, Chen CY, Wu RT. 1988. Gallotannins inhibit growth, water-
insoluble glucan synthesis, and aggregation of mutans streptococci. J
Dent Res 67:5155. https://doi.org/10.1177/00220345880670011001.
22. Nakahara K, Kawabata S, Ono H, Ogura K, Tanaka T, Ooshima T, Hamada S.
1993. Inhibitory effect of oolong tea polyphenols on glycosyltransferases of
mutans streptococci. Appl Environ Microbiol 59:968973. https://doi.org/10
.1128/aem.59.4.968-973.1993.
23. Wolinsky LE, Mania S, Nachnani S, Ling S. 1996. The inhibiting effect of
aqueous Azadirachta indica (Neem) extract upon bacterial properties
inuencing in vitro plaque formation. J Dent Res 75:816822. https://doi
.org/10.1177/00220345960750021301.
24. Koo H, Smith Vacca AM, Bowen WH, Rosalen PL, Cury JA, Park YK. 2000.
Effects of Apis mellifera propolis on the activities of streptococcal gluco-
syltransferases in solution and adsorbed onto saliva-coated hydroxyapa-
tite. Caries Res 34:418426. https://doi.org/10.1159/000016617.
25. Koo H, Seils J, Abranches J, Burne RA, Bowen WH, Quivey RG. 2006. Inu-
ence of apigenin on gtf gene expression in Streptococcus mutans UA159.
Antimicrob Agents Chemother 50:542546. https://doi.org/10.1128/AAC
.50.2.542-546.2006.
26. Kim H-S, Cha E, Kim Y, Jeon YH, Olson BH, Byun Y, Park H-D. 2016. Raf-
nose, a plant galactoside, inhibits Pseudomonas aeruginosa biolm forma-
tion via binding to LecA and decreasing cellular cyclic diguanylate levels.
Sci Rep 6:25318. https://doi.org/10.1038/srep25318.
27. Jovanovic-Malinovska R, Kuzmanova S, Winkelhausen E. 2014. Oligosaccharide
prole in fruits and vegetables as sources of prebiotics and functional foods. Int
J Food Prop 17:949965. https://doi.org/10.1080/10942912.2012.680221.
28. Ito K, Ito S, Shimamura T, Weyand S, Kawarasaki Y, Misaka T, Abe K,
Kobayashi T, Cameron AD, Iwata S. 2011. Crystal structure of glucansu-
crase from the dental caries pathogen Streptococcus mutans. J Mol Biol
408:177186. https://doi.org/10.1016/j.jmb.2011.02.028.
29. Forssten SD, Björklund M, Ouwehand AC. 2010. Streptococcus mutans,
caries and simulation models. Nutrients 2:290298. https://doi.org/10
.3390/nu2030290.
30. Li YF, Sun HW, Gao R, Liu KY, Zhang HQ, Fu QH, Qing SL, Guo G, Zou QM.
2015. Inhibited biolm formation and improved antibacterial activity of a
novel nanoemulsion against cariogenic Streptococcus mutans in vitro and
in vivo. Int J Nanomed 10:447462. https://doi.org/10.2147/IJN.S72920.
Inhibition of S. mutans Biolm Formation by Rafnose Microbiology Spectrum
May/June 2022 Volume 10 Issue 3 10.1128/spectrum.02076-21 12
31. Nagasawa R, Sato T, Senpuku H. 2017. Rafnose induces biolm forma-
tion by Streptococcus mutans in low concentrations of sucrose by increas-
ing production of extracellular DNA and fructan. Appl Environ Microbiol
83:e00869-17. https://doi.org/10.1128/AEM.00869-17.
32. Tamesada M, Kawabata S, Fujiwara T, Hamada S. 2004. Synergistic effects
of streptococcal glucosyltransferases on adhesive biolm formation. J
Dent Res 83:874879. https://doi.org/10.1177/154405910408301110.
33. Kawada-Matsuo M, Oogai Y, Komatsuzawa H. 2016. Sugar allocation to
metabolic pathways is tightly regulated and affects the virulence of Strep-
tococcus mutans. Genes 8:11. https://doi.org/10.3390/genes8010011.
34. Wunder D, Bowen WH. 1999. Action of agents on glucosyltransferases from
Streptococcus mutans in solution and adsorbed to experimental pellicle. Arch
Oral Biol 44:203214. https://doi.org/10.1016/s0003-9969(98)00129-0.
35. Vacca-Smith AM, Bowen WH. 1998. Binding properties of streptococcal glu-
cosyltransferases for hydroxyapatite, saliva-coated hydroxyapatite, and bac-
terial surfaces. Arch Oral Biol 43:103110. https://doi.org/10.1016/s0003
-9969(97)00111-8.
36. Ryu E-J, An S-J, Sim J, Sim J, Lee J, Choi B-K. 2020. Use of D-galactose to
regulate biolm growth of oral streptococci. Arch Oral Biol 111:104666.
https://doi.org/10.1016/j.archoralbio.2020.104666.
37. Aduse-Opoku J, Gilpin ML, Russell RR. 1989. Genetic and antigenic
comparison of Streptococcus mutans fructosyltransferase and glucan-
binding protein. FEMS Microbiol Lett 50:279282. https://doi.org/10
.1016/0378-1097(89)90432-1.
38. Russell R, Aduse-Opoku J, Sutcliffe I, Tao L, Ferretti J. 1992. A binding pro-
tein-dependent transport system in Streptococcus mutans responsible for
multiple sugar metabolism. J Biol Chem 267:46314637. https://doi.org/
10.1016/S0021-9258(18)42880-3.
39. Burne RA, Wen ZT, Chen YY, Penders JE. 1999. Regulation of expression of
the fructan hydrolase gene of Streptococcus mutans GS-5 by induction
and carbon catabolite repression. J Bacteriol 181:28632871. https://doi
.org/10.1128/JB.181.9.2863-2871.1999.
40. Heydorn A, Nielsen AT, Hentzer M, Sternberg C, Givskov M, Ersbøll BK, Molin
S. 2000. Quantication of biolm structures by the novel computer program
COMSTAT. Microbiology 146:23952407. https://doi.org/10.1099/00221287
-146-10-2395.
41. Decker E-M, Klein C, Schwindt D, Von Ohle C. 2014. Metabolic activity of
Streptococcus mutans biolms and gene expression during exposure to xyli-
tol and sucrose. Int J Oral Sci 6:195204. https://doi.org/10.1038/ijos.2014.38.
42. Rouabhia M, Semlali A. 2021. Electronic cigarette vapor increases Streptococ-
cus mutans growth, adhesion, biolm formation, and expression of the bio-
lm-associated genes. Oral Dis 27:639647. https://doi.org/10.1111/odi.13564.
43. Livak KJ, Schmittgen TD. 2001. Analysis of relative gene expression data
using real-time quantitative PCR and the 2
2DDCT
method. Methods 25:
402408. https://doi.org/10.1006/meth.2001.1262.
44. Ooshima T, Osaka Y, Sasaki H, Osawa K, Yasuda H, Matsumura M, Sobue S,
Matsumoto M. 2000. Caries inhibitory activity of cacao bean husk extract
in in-vitro and animal experiments. Arch Oral Biol 45:639645. https://doi
.org/10.1016/s0003-9969(00)00042-x.
45. Song J-H, Kim S-K, Chang K-W, Han S-K, Yi H-K, Jeon J-G. 2006. In vitro inhibi-
tory effects of Polygonum cuspidatum on bacterial viability and virulence fac-
tors of Streptococcus mutans and Streptococcus sobrinus.ArchOralBiol51:
11311140. https://doi.org/10.1016/j.archoralbio.2006.06.011.
Inhibition of S. mutans Biolm Formation by Rafnose Microbiology Spectrum
May/June 2022 Volume 10 Issue 3 10.1128/spectrum.02076-21 13
... Although non-nutritive sweeteners cannot be digested to generate energy, they can be potentially toxic to cells following cellular absorption [25]. Nevertheless, non-nutritive sweeteners have also been shown recently to be beneficial in managing microbial infections [26][27][28][29][30]. Bulk sweeteners also exhibit antibacterial efficacy against a variety of drug-resistant pathogens [31][32][33]. ...
... However, the mechanisms underlying the inhibition of oral biofilm formation and plaque reduction due to the use of sugar substitutes have not been fully elucidated. Bulk and non-nutritive sweeteners have been shown to reduce oral biofilm formation and activity by suppressing the expression of biofilm-and virulence-related genes (Fig. 3) [26][27][28]87]. In addition, unlike sucrose, bulk and non-nutritive sweeteners cannot be utilized by S. mutans as a substrate for biofilm matrix synthesis, resulting in reduced biofilm formation [32]. ...
... In particular, raffinose at 1 μM or higher was also shown to inhibit the formation of mixed biofilms of S. aureus and P. aeruginosa. In particular, raffinose was found to inhibit S. mutans biofilm (formed in a medium supplemented with 10 μM sucrose) formation and Gtf-related gene expression [28]. In addition, 10 % acesulfame-K and 7.5 % sucralose inhibited the biofilm formation of Porphyromonas gingivalis, an anaerobic periodontal pathogen, and showed bactericidal activity against bacteria within the biofilm [88]. ...
Article
Full-text available
There is a growing interest in using sweeteners for taste improvement in the food and drink industry. Sweeteners were found to regulate the formation or dispersal of structural components of microbial biofilms. Dietary sugars may enhance biofilm formation and facilitate the development of antimicrobial resistance, which has become a major health issue worldwide. In contrast, bulk and non-nutritive sweeteners are also beneficial for managing microbial infections. This review discusses the clinical significance of oral biofilms formed upon the administration of nutritive and non-nutritive sweeteners. The underlying mechanism of action of sweeteners in the regulation of mono- or poly-microbial biofilm formation and destruction is comprehensively discussed. Bulk and non-nutritive sweeteners have also been used in conjunction with antimicrobial substances to reduce microbial biofilm formation. Formulations with bulk and non-nutritive sweeteners have been demonstrated to be particularly efficient in this regard. Finally, future perspectives with respect to advancing our understanding of mechanisms underlying biofilm regulation activities of sweeteners are presented as well. Several alternative strategies for the application of bulk sweeteners and non-nutritive sweeteners have been employed to control the biofilm-forming microbial pathogens. Gaining insight into the underlying mechanisms responsible for enhancing or inhibiting biofilm formation and virulence properties by both mono- and poly-microbial species in the presence of the sweetener is crucial for developing a therapeutic agent to manage microbial infections.
... Dental caries is one of the three common oral diseases and an important global public health problem. The occurrence of dental caries affects the living quality of countless people around the world and brings a huge burden of healthcare costs (Ham et al. 2022). If dental caries is not treated in time, it can cause pulpitis, periodontitis, gingivitis, and eventual tooth loss (Durand et al. 2019). ...
... GtfC produces both water-insoluble glucan connected by α-1,3 glycosidic bonds and water-soluble glucan connected by α-1,6 glycosidic bonds, of which water-insoluble glucan is the main extracellular polysaccharide. GtfD uses sucrose to synthesize α-1,6-linked water-soluble glucan (Ham et al. 2022;. These glucosyltransferases work together to produce water-insoluble glucan, which facilitates bacterial adhesion and aggregation on tooth surfaces, while water-soluble glucan may provide a nutrient source for colonizing pathogens (Jakubovics et al., 2021;Kong et al. 2021; Traditional methods of removing dental plaque mainly include mechanical cleaning and the use of antibacterial agents. ...
Article
Aims: To solve the shortcomings of poor solubility, easy volatilization and decomposition, propolis essential oil microemulsion (PEOME) was prepared. The antibacterial, anti-biofilm activities and action mechanism of PEOME against Streptococcus mutans was analyzed. Methods: PEOME was prepared using anhydrous ethanol and Tween-80 as the cosurfactant and surfactant respectively. The antibacterial activity of PEOME against Streptococcus mutans was evaluated using the agar disk-diffusion method and broth microdilution method. The effects of PEOME on S. mutans biofilm was detected through the assays of crystal violet (CV), XTT reduction, lactic dehydrogenase (LDH) and calcium ions leaking, live/dead staining and scanning electron microscopy (SEM). And the anti-biofilm mechanism of PEOME was elaborated by the assays of extracellular polysaccharide (EPS) production and glucosyltransferase (GTF) activity. Results: The inhibition zone diameter (DIZ) of PEOME against S. mutans was 31 mm, while the minimal inhibitory concentration (MIC) was 2.5 µL mL-1. CV and XTT assays showed that PEOME could prevent fresh biofilm formation and disrupt preformed biofilm through decreasing the activities and biomass of biofilm. The leaking assays for LDH and calcium ions, as well as the live/dead staining assay, indicated that PEOME was able to damage the integrity of bacterial cell membranes within the biofilm. SEM revealed that PEOME had a noticeable inhibitory effect on bacterial adhesion and aggregation through observing the overall structure of biofilm. The assays of EPS production and GTF activity suggested that PEOME could reduce EPS production by inhibiting the activity of GTFs, thus showing an anti-biofilm effect. Conclusions: The significant antibacterial and anti-biofilm activities against S. mutans of PEOME meant that PEOME has great potential to be developed as a drug to prevent and cure dental caries caused by S. mutans.
... In addition, benzoic acid is associated with vigorous antibacterial activity by inhibiting the proliferation of bacteria such as Escherichia coli and Bacillus subtilis [93]. In contrast, raffinose-related metabolites inhibit Streptococcus [94], and these metabolites were found to be upregulated in our metabolome analysis. ...
Article
Full-text available
Curcumin, a polyphenol derived from Curcuma longa, used as a dietary spice, has garnered attention for its therapeutic potential, including antioxidant, anti-inflammatory, and antimicrobial properties. Despite its known benefits, the precise mechanisms underlying curcumin’s effects on consumers remain unclear. To address this gap, we employed the genetic model Drosophila melanogaster and leveraged two omics tools—transcriptomics and metabolomics. Our investigation revealed alterations in 1043 genes and 73 metabolites upon supplementing curcumin into the diet. Notably, we observed genetic modulation in pathways related to antioxidants, carbohydrates, and lipids, as well as genes associated with gustatory perception and reproductive processes. Metabolites implicated in carbohydrate metabolism, amino acid biosynthesis, and biomarkers linked to the prevention of neurodegenerative diseases such as schizophrenia, Alzheimer’s, and aging were also identified. The study highlighted a strong correlation between the curcumin diet, antioxidant mechanisms, and amino acid metabolism. Conversely, a lower correlation was observed between carbohydrate metabolism and cholesterol biosynthesis. This research highlights the impact of curcumin on the diet, influencing perception, fertility, and molecular wellness. Furthermore, it directs future studies toward a more focused exploration of the specific effects of curcumin consumption.
... The raffinose contents in Samples A, B, and C were 2.14 g/100g, 2.75 g/100g, and 1.49 g/100g, respectively, indicating that Samples A and B contained approximately 1.5 times more raffinose than Sample C. Allose is generally known for its antioxidant [24], anti-inflammatory [25], and antitumor effects [26]. Raffinose has been reported to balance the intestinal microflora in humans [12,13] and inhibit biofilm formation by the Streptococcus species in the human oral cavity [27]. Thus, allose and raffinose have health benefits. ...
Article
Full-text available
We investigated the physicochemical properties of Japanese rice wines, including their functional properties and carbohydrate and amino acid content in solution and solid state. Three samples were tested. The glucose, allose, and raffinose contents in samples (A, B, C) in g/100 g were (3.47, 3.45, 7.05), (1.60, 1.63, 1.61), and (2.14, 2.75, 1.49), respectively. The total amino acid in µmol/mL was (3.1, 3.5, 4.4). Glutamic acid, alanine, and arginine varied in content across the samples. The viscosity (10 °C) and activation energy (ΔE) calculated using the Andrade equation were (2.81 ± 0.03, 2.74 ± 0.06, 2.69 ± 0.03) mPa-s and (22.3 ± 1.1, 22.0 ± 0.2, 21.3 ± 0.5) kJ/mol, respectively. Principal component analysis using FT-IR spectra confirmed the separation of the samples into principal components 2 and 3. The IC50 values from the DPPH radical scavenging test were (2364.7 ± 185.3, 3041.9 ± 355.1, 3842.7 ± 228.1) µg/mL. Thus, the three rice wines had different carbohydrate and amino acid contents, viscosities, and antioxidant capacities.
... To study the effect of CA@CS NC on the expression of genes of S. mutans, we contrasted the effects of CA@CS NC and blank groups on the genes of S. mutans at equal concentrations. Among the genes studied (Table 1), there are 5 genes involved in QS regulation (comB, comE, comS, comA and comR), 1 gene involved in biosynthesis and adhesion (gbpB), 1 gene involved in two-component signal transduction system and bacterial virulence (vicR), 3 genes involved in extracellular polysaccharide synthesis (gtfB, gtfC, gtfD) [26,[30][31][32][33]. The organism was treated with CA@CS NC and cultured in medium for 12 h. ...
Article
Full-text available
Background Dental caries is a multifactorial disease, and the bacteria such as Streptococcus mutans (S. mutans) is one of the risk factors. The poor effect of existing anti-bacterial is mainly related to drug resistance, the short time of drug action, and biofilm formation. Methods To address this concern, we report here on the cinnamaldehyde (CA) loaded chitosan (CS) nanocapsules (CA@CS NC) sustained release CA for antibacterial treatment. The size, ζ-potential, and morphology were characterized. The antibacterial activities in vitro were studied by growth curve assay, pH drop assay, biofilm assay, and qRT-PCR In addition, cytotoxicity assay, organ index, body weight, and histopathology results were analyzed to evaluate the safety and biocompatibility in a rat model. Results CA@CS NC can adsorb the bacterial membrane due to electronic interaction, releasing CA slowly for a long time. At the same time, it has reliable antibacterial activity against S. mutans and downregulated the expression levels of QS, virulence, biofilm, and adhesion genes. In addition, it greatly reduced the cytotoxicity of CA and significantly inhibited dental caries in rats without obvious toxicity. Conclusion Our results showed that CA@CS NC had antibacterial and antibiofilm effects on S. mutans and inhibit dental caries. Besides, it showed stronger efficacy and less toxicity, and was able to adsorb bacteria releasing CA slowly, providing a new nanomaterial solution for the treatment of dental caries.
... Sucrose stimulates the expression of gtfBC as does interaction with other oral microbiota members (e.g., C. albicans). However, evidence recently reported suggests that treatment with the trisaccharide raffinose negatively affects Gtf activity and gtf gene expression and biofilm development (Ham et al., 2022), although the mechanisms operating in raffinose-dependent S. mutans biofilm inhibition are not known. ...
Article
Full-text available
It is widely acknowledged that the human‐associated microbial community influences host physiology, systemic health, disease progression, and even behavior. There is currently an increased interest in the oral microbiome, which occupies the entryway to much of what the human initially encounters from the environment. In addition to the dental pathology that results from a dysbiotic microbiome, microbial activity within the oral cavity exerts significant systemic effects. The composition and activity of the oral microbiome is influenced by (1) host–microbial interactions, (2) the emergence of niche‐specific microbial “ecotypes,” and (3) numerous microbe–microbe interactions, shaping the underlying microbial metabolic landscape. The oral streptococci are central players in the microbial activity ongoing in the oral cavity, due to their abundance and prevalence in the oral environment and the many interspecies interactions in which they participate. Streptococci are major determinants of a healthy homeostatic oral environment. The metabolic activities of oral Streptococci, particularly the metabolism involved in energy generation and regeneration of oxidative resources vary among the species and are important factors in niche‐specific adaptations and intra‐microbiome interactions. Here we summarize key differences among streptococcal central metabolic networks and species‐specific differences in how the key glycolytic intermediates are utilized.
... In vitro culturing method [116] Raffinose P. aeruginosa, S. aureus It effectively reduces the biofilm formation of tested pathogens in a dose-dependent manner ...
Article
Full-text available
Antimicrobial resistance (AMR) is a significant public health concern worldwide. The continuous use and misuse of antimicrobial agents have led to the emergence and spread of resistant strains of bacteria, which can cause severe infections that are difficult to treat. One of the reasons for the constant development of new antimicrobial agents is the need to overcome the resistance that has developed against existing drugs. However, this approach is not sustainable in the long term, as bacteria can quickly develop resistance to new drugs as well. Additionally, the development of new drugs is costly and time-consuming, and there is no guarantee that new drugs will be effective or safe. An alternative approach to combat AMR is to focus on improving the body’s natural defenses against infections by using probiotics, prebiotics, and synbiotics, which are helpful to restore and maintain a healthy balance of bacteria in the body. Probiotics are live microorganisms that can be consumed as food or supplements to promote gut health and improve the body’s natural defenses against infections. Prebiotics are non-digestible fibers that stimulate the growth of beneficial bacteria in the gut, while synbiotics are a combination of probiotics and prebiotics that work together to improve gut health. By promoting a healthy balance of bacteria in the body, these can help to reduce the risk of infections and the need for antimicrobial agents. Additionally, these approaches are generally safe and well tolerated, and they do not contribute to the development of AMR. In conclusion, the continuous development of new antimicrobial agents is not a sustainable approach to combat AMR. Instead, alternative approaches such as probiotics, prebiotics, and synbiotics should be considered as they can help to promote a healthy balance of bacteria in the body and reduce the need for antibiotics.
Article
Full-text available
Cassava is an ideal food security crop in marginal and drought environment. However, the post-harvest storage of cassava is urgent problem to be resolved. In this study, the storage tolerant and non-tolerant cassava were screened by measuring the change of Peroxidase (POD), Superoxide dismutase (SOD), Catalase (CAT) and Malondialdehyde (MDA) in seven cultivars of cassava. Compared with other cultivars, the cultivar of SC14 showed the highest level of SOD, MDA and POD respectively at 0 day, 12 day and 9 day postharvest while exhibited lowest level of CAT at 0 day postharvest, indicating the strongest antioxidant capability and storage tolerance. In contrast, GR15231, termed as storage non-tolerance cultivars, showed lowest SOD and POD at 12 day and kept a relative high level of CAT at 12 day post-harvest. In addition, SC14 has higher level of starch and dry substance than GR15231. Mass spectrum was performed for SC14 and GR15231 to explore the key metabolites regulating the storage tolerance of cassava. The results showed that the expression of glutathione (reduced) and raffinose was significantly decreased at 12 day post-harvest both in tolerant SC14 and non-tolerant GR15231. Compared with GR15231, SC14 showed higher level of raffinose both at 0 and 12 day post-harvest, indicating that raffinose may be the potential metabolites protecting SC14 cultivar from deterioration post-harvest. Additionally, raffinose ratio of SC14a/SC14b was five times less than that of GR15231a/GR15231b, reflecting the slower degradation of raffinose in SC14 cultivar compared with GR15231 cultivar. In conclusion, the antioxidant microenvironment induced by reduced glutathione and higher level of raffinose in SC14 cultivar might be the promising metabolites to improve its antioxidant capacity and antibiosis and thus maintained the quality of Cassava root tubers.
Article
In this study, the influences of Lactobacillus plantarum JHT78 fermentation on the physiological properties, antioxidant activities, and volatile/non-volatile metabolites of watermelon juices were comprehensively investigated. The results indicated that total polyphenols flavonoids and anthocyanin in the watermelon juices remarkably increased through L. plantarum JHT78 fermentation. L. plantarum JHT78 fermentation enhanced the antioxidant activities, lipase inhibition, and α-glucosidase activities of watermelon juices. A total of 62 volatile compounds were detected using HS-SPME-GC-MS, mainly including 11 acids, 8 aldehydes, 7 ketones, and 7 alcohols. The abundance of 19 volatile compounds especially for acids remarkably increased for the fermentated watermelon juice. Furthermore, non-volatile compounds detected by UHPLC-QTOF-MS revealed that L. plantarum JHT78 significantly altered the non-volatile compounds of watermelon juices, especially increased indole-3-lactic acid. The results confirmed that L. plantarum JHT78 enhanced the functionality of watermelon juices thus providing a theoretical basis for the development of LAB on plant-based beverages.
Article
Full-text available
In some diseases, a very important role is played by the ability of bacteria to form multi-dimensional complex structure known as biofilm. The most common disease of the oral cavity, known as dental caries, is a top leader. Streptococcus mutans, one of the many etiological factors of dental caries, is a microorganism which is able to acquire new properties allowing for the expression of pathogenicity determinants determining its virulence in specific environmental conditions. Through the mechanism of adhesion to a solid surface, S. mutans is capable of colonizing the oral cavity and also of forming bacterial biofilm. Additional properties enabling S. mutans to colonize the oral cavity include the ability to survive in an acidic environment and specific interaction with other microorganisms colonizing this ecosystem. This review is an attempt to establish which characteristics associated with biofilm formation--virulence determinants of S. mutans--are responsible for the development of dental caries. In order to extend the knowledge of the nature of Streptococcus infections, an attempt to face the following problems will be made: Biofilm formation as a complex process of protein-bacterium interaction. To what extent do microorganisms of the cariogenic flora exemplified by S. mutans differ in virulence determinants "expression" from microorganisms of physiological flora? How does the environment of the oral cavity and its microorganisms affect the biofilm formation of dominant species? How do selected inhibitors affect the biofilm formation of cariogenic microorganisms?
Article
Full-text available
For decades, fluoride has been used extensively as an anti-caries agent. It not only protects dental hard tissue, but also inhibits bacterial growth and metabolism. The antimicrobial action of fluoride is shown in three main aspects: the acidogenicity, acidurance, and adherence to the tooth surface. To counteract the toxic effect of fluoride, oral bacteria are able to develop resistance to fluoride through either phenotypic adaptation or genotypic changes. Strains that acquire fluoride resistance through the latter route show stable resistance and can usually resist much higher fluoride levels than the corresponding wild-type strain. This review summarizes the characteristics of fluoride-resistant strains and explores the mechanisms of fluoride resistance, in particular the recent discovery of the fluoride exporters. Since the fluoride resistance of the cariogenic bacterium Streptococcus mutans has been studied most extensively, this review mainly discusses the findings related to this species.
Article
Full-text available
Streptococcus mutans is the primary etiological agent of dental caries and causes tooth decay by forming a firmly attached biofilm on tooth surfaces. Biofilm formation is induced by the presence of sucrose, which is a substrate for the synthesis of extracellular polysaccharides but not in the presence of oligosaccharides. Nonetheless, in this study, we found that raffinose, which is an oligosaccharide with an intestinal regulatory function and antiallergic effect, induced biofilm formation by S. mutans in a mixed culture with sucrose, which was at concentrations less than those required to induce biofilm formation directly. We analyzed the possible mechanism behind the small requirement for sucrose for biofilm formation in the presence of raffinose. Our results suggested that sucrose contributed to an increase in bacterial cell surface hydrophobicity and biofilm formation. Next, we examined how the effects of raffinose interacted with the effects of sucrose for biofilm formation. We showed that the presence of raffinose induced fructan synthesis by fructosyltransferase and aggregated extracellular DNA (eDNA, which is probably genomic DNA released from dead cells) into the biofilm. eDNA seemed to be important for biofilm formation, because the degradation of DNA by DNase I resulted in a significant reduction in biofilm formation. When assessing the role of fructan in biofilm formation, we found that fructan enhanced eDNA-dependent cell aggregation. Therefore, our results show that raffinose and sucrose have cooperative effects and that this induction of biofilm formation depends on supportive elements that mainly consist of eDNA and fructan. IMPORTANCE The sucrose-dependent mechanism of biofilm formation in Streptococcus mutans has been studied extensively. Nonetheless, the effects of carbohydrates other than sucrose are inadequately understood. Our findings concerning raffinose advance the understanding of the mechanism underlying the joint effects of sucrose and other carbohydrates on biofilm formation. Since raffinose has been reported to have positive effects on enterobacterial flora, research on the effects of raffinose on the oral flora are required prior to its use as a beneficial sugar for human health. Here, we showed that raffinose induced biofilm formation by S. mutans in low concentrations of sucrose. The induction of biofilm formation generally generates negative effects on the oral flora. Therefore, we believe that this finding will aid in the development of more effective oral care techniques to maintain oral flora health.
Article
Full-text available
Bacteria take up and metabolize sugar as a carbohydrate source for survival. Most bacteria can utilize many sugars, including glucose, sucrose, and galactose, as well as amino sugars, such as glucosamine and N-acetylglucosamine. After entering the cytoplasm, the sugars are mainly allocated to the glycolysis pathway (energy production) and to various bacterial component biosynthesis pathways, including the cell wall, nucleic acids and amino acids. Sugars are also utilized to produce several virulence factors, such as capsule and lipoteichoic acid. Glutamine-fructose-6-phosphate aminotransferase (GlmS) and glucosamine-6-phosphate deaminase (NagB) have crucial roles in sugar distribution to the glycolysis pathway and to cell wall biosynthesis. In Streptococcus mutans, a cariogenic pathogen, the expression levels of glmS and nagB are coordinately regulated in response to the presence or absence of amino sugars. In addition, the disruption of this regulation affects the virulence of S. mutans. The expression of nagB and glmS is regulated by NagR in S. mutans, but the precise mechanism underlying glmS regulation is not clear. In Staphylococcus aureus and Bacillus subtilis, the mRNA of glmS has ribozyme activity and undergoes self-degradation at the mRNA level. However, there is no ribozyme activity region on glmS mRNA in S. mutans. In this review article, we summarize the sugar distribution, particularly the coordinated regulation of GlmS and NagB expression, and its relationship with the virulence of S. mutans.
Article
Full-text available
Biofilm formation on biotic or abiotic surfaces has unwanted consequences in medical, clinical, and industrial settings. Treatments with antibiotics or biocides are often ineffective in eradicating biofilms. Promising alternatives to conventional agents are biofilm-inhibiting compounds regulating biofilm development without toxicity to growth. Here, we screened a biofilm inhibitor, raffinose, derived from ginger. Raffinose, a galactotrisaccharide, showed efficient biofilm inhibition of Pseudomonas aeruginosa without impairing its growth. Raffinose also affected various phenotypes such as colony morphology, matrix formation, and swarming motility. Binding of raffinose to a carbohydrate-binding protein called LecA was the cause of biofilm inhibition and altered phenotypes. Furthermore, raffinose reduced the concentration of the second messenger, cyclic diguanylate (c-di-GMP), by increased activity of a c-di-GMP specific phosphodiesterase. The ability of raffinose to inhibit P. aeruginosa biofilm formation and its molecular mechanism opens new possibilities for pharmacological and industrial applications.
Article
Full-text available
Glycosyltransferase (Gtf) is one of the crucial virulence factors of Streptococcus mutans, a major etiological pathogen of dental caries. All the available evidence indicates that extracellular polysaccharide, particularly glucans produced by S. mutans Gtfs, contribute to the cariogenicity of dental biofilms. Therefore, inhibition of Gtf activity and the consequential polysaccharide synthesis may impair the virulence of cariogenic biofilms, which could be an alternative strategy to prevent the biofilm-related disease. Up to now, many Gtf inhibitors have been recognized in natural products, which remain the major and largely unexplored source of Gtf inhibitors. These include catechin-based polyphenols, flavonoids, proanthocyanidin oligomers, polymeric polyphenols, and some other plant-derived compounds. Metal ions, oxidizing agents, and some other synthetic compounds represent another source of Gtf inhibitors, with some novel molecules either discovered by structure-based virtual screening or synthesized based on key structures of known inhibitors as templates. Antibodies that inhibit one or more Gtfs have also been developed as topical agents. Although many agents have been shown to possess potent inhibitory activity against glucan synthesis by Gtfs, bacterial cell adherence, and caries development in animal models, much research remains to be performed to find out their mechanism of action, biological safety, cariostatic efficacies, and overall influence on the entire oral community. As a strategy to inhibit the virulence of cariogenic microbes rather than eradicate them from the microbial community, Gtf inhibition represents an approach of great potential to prevent dental caries.
Article
Full-text available
The aim of this study was to prepare a novel nanoemulsion loaded with poorly water-soluble chlorhexidine acetate (CNE) to improve its solubility, and specifically enhance the antimicrobial activity against Streptococcus mutans in vitro and in vivo. In this study, a novel CNE nanoemulsion with an average size of 63.13 nm and zeta potential of −67.13 mV comprising 0.5% CNE, 19.2% Tween 80, 4.8% propylene glycol, and 6% isopropyl myristate was prepared by the phase inversion method. Important characteristics such as the content, size, zeta potential, and pH value of CNE did not change markedly, stored at room temperature for 1 year. Also, compared with chlorhexidine acetate water solution (CHX), the release profile results show that the CNE has visibly delayed releasing effect in both phosphate-buffered saline and artificial saliva solutions (P<0.005). The minimum inhibitory concentration and minimum bactericidal concentration of CHX for S. mutans (both 0.8 μg/mL) are both two times those of CNE (0.4 μg/mL). Besides, CNE of 0.8 μg/mL exhibited fast-acting bactericidal efficacy against S. mutans, causing 95.07% death within 5 minutes, compared to CHX (73.33%) (P<0.01). We observed that 5 mg/mL and 2 mg/mL CNE were both superior to CHX, significantly reducing oral S. mutans numbers and reducing the severity of carious lesions in Sprague Dawley rats (P<0.05), in an in vivo test. CNE treatment at a concentration of 0.2 μg/mL inhibited biofilm formation more effectively than CHX, as indicated by the crystal violet staining method, scanning electron microscopy, and atomic force microscopy. The cell membrane of S. mutans was also severely disrupted by 0.2 μg/mL CNE, as indicated by transmission electron microscopy. These results demonstrated that CNE greatly improved the solubility and antimicrobial activity of this agent against S. mutans both in vitro and in vivo. This novel nanoemulsion is a promising medicine for preventing and curing dental caries.
Article
Objective It still not known whether electronic cigarettes (e‐cigarettes) contribute to dental caries. This study aimed to evaluate the effect of e‐cigarettes on the growth of Streptococcus mutans , the formation of biofilm, and the expression of certain virulence genes. Materials and Methods S. mutans cells were exposed or not to e‐cigarettes with and without nicotine or to cigarette smoke twice a day for 15 min each exposure period. The bacterial growth and the expression of glucosyltranferase, competence, and glucan‐binding genes were evaluated after 24 h. Biofilm formation was assessed after 1, 2, and 3 days. S. mutans adhesion and growth to e‐cigarette exposed human teeth were assessed. Results We observed an increase in S. mutans growth with e‐cigarettes, mainly at the early culture period. This was confirmed by an increase of biofilm mass ranging from 8 ± 0.5 mg with the control to 47 ± 5 mg after six exposures to nicotine‐rich e‐cigarettes. S. mutans cells adhered better to e‐cigarette exposed teeth. E‐cigarettes increased the expression of glucosyltranferase , competence, and glucan‐binding genes. Conclusions E‐cigarettes increased the growth of S. mutans , and the expression of virulent genes. E‐cigarettes promoted the adhesion to, and formation of biofilms on teeth surfaces.
Article
In the oral microbial community, commensals can compete with pathogens and reduce their colonization in the oral cavity. A substance that can inhibit harmful bacteria and enrich beneficial bacteria is required to maintain oral health. The purpose of this study was to examine the effect of d-galactose on the biofilm formation of the cariogenic bacteria Streptococcus mutans and oral commensal streptococci and to evaluate their use in solution and in paste form. Biofilms of S. mutans, Streptococcus oralis, and Streptococcus mitis were formed on saliva-coated glass slips in the absence or presence of d-galactose and evaluated by staining with 1 % crystal violet. d-Galactose significantly inhibited the biofilm formation of S. mutans at concentrations ranging from 2 μM to 200 mM but increased the biofilm formation of S. oralis and S. mitis at concentrations of 2-200 mM. d-Galactose significantly inhibited three glucosyltransferase genes, gtfB, gtfC, and gtfD. The effect of d-galactose in the form of solution and paste was evaluated using bovine teeth. Pretreatment with 100 mM d-galactose on bovine teeth resulted in significantly reduced S. mutans biofilm formation. Our results suggest that d-galactose can be a candidate substance for the development of oral hygiene products to prevent caries by inhibiting the biofilm formation of S. mutans and simultaneously increasing the biofilm formation of commensal oral streptococci.