Вы находитесь на странице: 1из 7

Original article 239

Subgingival microflora in inflammatory bowel disease


patients with untreated periodontitis
Fernanda Britoa, Cyrla Zaltmanc, Ana T.P. Carvalhob, Ricardo G. Fischera,
Rutger Perssond,e, Anders Gustafssonf and Carlos M.S. Figueredoa,f

Objective To analyze the subgingival microflora and S. mutans compared with controls both at gingivitis
composition of inflammatory bowel disease (IBD) patients and at periodontitis sites (P < 0.05). UC patients harbored
with untreated chronic periodontitis and compare them higher levels of S. aureus (P = 0.01) and P. anaerobius
with systemically healthy controls also having untreated (P = 0.05) than controls only in gingivitis sites.
chronic periodontitis.
Conclusion Our study showed that even with similar
Method Thirty IBD patients [15 with Crohn’s disease (CD) clinical periodontal parameters, IBD patients harbor higher
and 15 with ulcerative colitis (UC)] and 15 control levels of bacteria that are related to opportunistic
individuals participated in the study. All patients had been infections in inflamed subgingival sites that might be
diagnosed with untreated chronic periodontitis. From every harmful for the crucial microbe–host interaction. Eur J
patient, subgingival plaque was collected from four Gastroenterol Hepatol 25:239–245 c 2013 Wolters Kluwer
gingivitis and four periodontitis sites with paper points. Health | Lippincott Williams & Wilkins.
Samples from the same category (gingivitis or European Journal of Gastroenterology & Hepatology 2013, 25:239–245
periodontitis) in each patient were pooled together
and stored at – 708C until analysis using a checkerboard Keywords: checkerboard DNA–DNA hybridization, inflammatory bowel
disease, periodontal disease, subgingival plaque
DNA–DNA hybridization technique for 74 bacterial species.
a
Departament of Periodontology, Faculty of Odontology, bDepartment
Results Multiple-comparison analysis showed that the of Gastroenterology, Faculty of Medicine, Rio de Janeiro State University,
c
Department of Gastroenterology, Faculty of Medicine, Federal University of Rio
groups differed in bacterial counts for Bacteroides de Janeiro, Rio de Janeiro, Brazil, dDepartment of Periodontology, School of
ureolyticus, Campylobacter gracilis, Parvimonas micra, Dental Medicine, University of Bern, Bern, Switzerland, eDepartment of Oral
Medicine, School of Dentistry, University of Washington, Seattle, Washington,
Prevotella melaninogenica, Peptostreptococcus anaerobius, USA and fDepartment of Dental Medicine, Division of Perisodontology, Karolinska
Staphylococcus aureus, Streptococcus anginosus, Institutet, Stockholm, Sweden
Streptococcus intermedius, Streptococcus mitis, Correspondence to Carlos M.S. Figueredo, PhD, Departament of Periodontology,
Streptococcus mutans, and Treponema denticola Faculty of Odontology, Rio de Janeiro State University, Boulevard 28
de Setembro 157, Pavilhão de Pesquisa, Vila Isabel, Rio de Janeiro
(P < 0.001). CD patients had significantly higher levels 20551-030, Brazil
of these bacteria than UC patients either in gingivitis Tel/fax: + 55 212 868 8642; e-mail: cmfigueredo@hotmail.com
or in periodontitis sites (P < 0.05). CD patients harbored
Received 12 July 2012 Accepted 5 September 2012
higher levels of P. melaninogenica, S. aureus, S. anginosus,

Introduction thus, prevention of gingivitis is the primary preventive


Periodontal diseases are chronic inflammatory conditions measure for preventing periodontitis [3].
affecting the supporting structures of the teeth, which Evidence suggests that the effect of periodontitis might
might be divided into gingivitis and periodontitis. Plaque- not be limited just to the oral cavity, but it might have
induced gingivitis is an inflammation of the gums systemic consequences [4]. Periodontitis can elicit a
resulting from bacteria located at the gingival margin. systemic inflammatory response by activating the hepatic
The common clinical findings of plaque-induced gingivi- acute-phase response [5], and might represent one
tis include erythema, edema, bleeding, sensitivity, distant source of low-grade systemic inflammation.
tenderness, and enlargement. In gingivitis, there is no For this reason, periodontitis is significantly associated with
loss of supporting structures [1]. However, periodontitis several systemic conditions, including myocardial infarction,
involves the destruction of the periodontal ligament, stroke, diabetes, and preterm low birth weight [4].
bone, and gingival tissues, and can lead to tooth loss. The
inflammatory and immune responses in the gingival Studies have reported a high prevalence of periodontitis
pocket of periodontal patients are presumed to be in patients with inflammatory bowel disease (IBD) [6,7].
initiated and perpetuated by several bacterial species Our group found significantly more patients with period-
residing in a biofilm on the tooth surfaces referred to as ontitis in ulcerative colitis (UC) (90.0%) and Crohn’s
dental plaque [2]. Destructive periodontal disease is disease (CD) (81.8%) than controls (67.6%) [8]. Period-
a consequence of the interaction of genetic, environmental, ontal pockets can harbor a total estimate of 415 species in
host, and microbial factors. Periodontitis follows gingivitis; the subgingival plaque [9], and therefore, are appropriate
0954-691X
c 2013 Wolters Kluwer Health | Lippincott Williams & Wilkins DOI: 10.1097/MEG.0b013e32835a2b70

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
240 European Journal of Gastroenterology & Hepatology 2013, Vol 25 No 2

ecological niches for hosting microorganisms that could Rio de Janeiro State University and at the Clementino
act as opportunistic pathogens [10]. Guidelines for the Fraga Filho University Hospital of Federal University of
prevention of opportunistic infections in IBD patients Rio de Janeiro, both in Rio de Janeiro, Brazil. Employees
mention that the dental status needs to be evaluated and at both University Hospitals, and who did not show any
appropriate dental care has to be performed. However, no clinical signs of present systemic disease other than
special mention is made of the periodontal condition periodontitis, and who had not taken antibiotics, or anti-
of these patients [11]. inflammatory drugs for at least 6 months before the study,
were included in the control group. The smoking habit
Some attempts to identify the oral IBD microflora have
was registered as current smokers (those who currently
been made through the years. van Dyke et al. [12] studied
smoke cigarettes daily) and nonsmokers (those who had
the periodontal flora of IBD patients with and without
never smoked cigarettes or former smokers who had quit
periodontitis and found a unique microflora composed
smoking for >5 years).
predominantly of small, motile, Gram-negative rods that
were most consistent with the genus Wolinella. Sundh The diagnosis of CD or UC had been established
et al. [13] reported that CD patients had higher counts of previously by clinical, radiological, endoscopic, and
Streptococcus mutans than controls. More recently, Docktor histological analyses. Crohn’s Disease Activity Index [15]
et al. [14] found a significant decrease in overall diversity was used to assess disease activity in CD patients,
in the oral microbiome of pediatric CD. According to the whereas Truelove and Witts’ index [16] was used to
authors, further studies of the oral microbiome in IBD assess disease activity in UC patients. CD patients were
may be of potential diagnostic and prognostic value. taking immunomodulators (n = 7), aminosalicylates (n = 4),
and immunomodulators + aminosalicylates (n = 2). Two
The host–microbe interaction has been implicated in the
CD patients were not taking any medication. In the UC
pathogenesis of IBD in genetically susceptible hosts, but
group, the disease was active in three patients and was in
limited information exists about oral microbes in such
remission in 12 of the patients. In the UC group, patients
patients. Our hypothesis is that the presence of
were taking immunomodulators (n = 1), aminosalicylates
pathogenic bacteria in the inflamed subgingival area
(n = 9), and immunomodulators + aminosalicylates (n = 5).
might be harmful for the crucial microbe–host interaction
None of the individuals in the control group used any
in IBD patients. Therefore, our aim was to analyze the
prescribed medications.
subgingival microflora composition of IBD patients with
untreated chronic periodontitis and to compare with
systemically healthy controls also having untreated Clinical examination
chronic periodontitis. Clinical parameters were evaluated at six sites in all
teeth, excluding third molars. The parameters included
Materials and methods PPD, CAL, presence of plaque, and presence of bleeding
Selection of participants on probing (BOP). PPD, CAL, and BOP were measured
The Committee on Ethics and Research of the University using a conventional periodontal probe (Hu-Friedy,
Hospitals Pedro Ernesto and Clementino Fraga Filho, and Chicago, Illinois, USA). After the diagnosis of chronic
at the Karolinska Institutet (Stockholm, Sweden), periodontitis was made, the sites to be sampled were
approved the study. All the selected patients signed an selected. All the patients had moderate to severe chronic
informed consent. periodontitis [17].

Forty-five patients with untreated chronic periodontitis


Microbiological sampling
were enrolled in the present study. Thirty of these
Before sampling, supragingival plaque was removed and
patients were diagnosed with IBD: 15 had CD (seven
the gingival margins were wiped dry with a sterile cotton
women and eight men, mean age 38.2±11.4 years) and 15
pellet. Samples were taken from inflamed sites: four
had UC (eight women and seven men, mean age
gingivitis sites (PPD r 3 mm) and four periodontitis sites
45.0±10.5 years). Fifteen controls (eight women and
(PPD Z 5 mm). The sites were sampled from different
seven men, mean age 42.1±7.8 years) were examined for
teeth. The paper point was inserted into the gingival
comparisons. These individuals represent a subset sample
crevice for 5 s. The paper points from the same category
population of a previous study [8]. The patients
(gingivitis or periodontitis) in each individual were
previously diagnosed with periodontitis were invited to
pooled together and were stored in Eppendorf vials at
return for microbiological sampling. Among the patients
– 701C until analyzed with respect to microbiology.
who returned, those who presented at least five inflamed
To each vial with a subgingival bacterial sample, 0.15 ml
sites with probing pocket depths (PPDs) of at least 5 mm
Tris-EDTA buffer (10 mmol/l Tris-HCL, 1.0 mmol/l
and clinical attachment loss (CAL) of at least 3 mm
EDTA, pH 7.6) was added to each RNase-free, DNase-
in different teeth were selected for the present study.
free, DNA-free, and pyrogen-free sterile 1.5 ml natural flat-
CD and UC patients were outpatients attending to the cap microcentrifuge vials (Starlab GmbH, Ahrensburg,
IBD clinics at Pedro Ernesto University Hospital of the Germany).

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Subgingival flora in IBD patients Brito et al. 241

Microbiological processing Statistical analysis


Samples were analyzed using the checkerboard DNA– The Kolmogorov–Smirnov test was used to assess the
DNA hybridization technique and 74 bacterial species data distribution for each variable presented. A normal
were assayed (Table 1) as described elsewhere [18,19]. distribution curve was found for age, the number of
At the laboratory, 0.10 ml 0.5 mol/l NaOH was added to remaining teeth, percentage plaque, mean PPD and PPD
each vial. Bacterial DNA was extracted, concentrated on for sites from which the bacterial samples were collected,
nylon membranes, and fixed by cross-linking using mean CAL, and CAL for sites from which the bacterial
ultraviolet light. The membranes with fixed DNA were samples were collected, and for the number of months
placed in a Miniblotter 45 (Roche Diagnostics GmbH, with a diagnosis of CD or UC. All other study variables
Mannheim, Germany). Signals were detected by fluores- were identified as having non-normal distribution curves.
cence using the Storm Fluor-Imager (Stratalinker 1800; Multivariate analysis (post-hoc Bonferroni), Kruskal–
Stratagene, La Jolla, California, USA) with a setup of Wallis analysis of variance (ANOVA), as well as repeated
200 mm and 600 V. The digitized information was analyzed independent t-tests, and Mann–Whitney U-tests were
using a software program (ImageQuant; Amersham used as appropriate for data with a normal distribution or
Pharmacia, Piscataway, New Jersey, USA) allowing a not. For the microbiological data, significance was set at
comparison of the density of 19 sample lanes against P less than 0.001. Otherwise, P less than 0.05 was used to
the two standard lanes (105 or 106 cells). Signals were indicate significance. The bacteria shown in the result
converted into absolute counts by comparisons with these section are those that consistently showed agreement by
standards. Patient-based mean values for the bacterial one-way ANOVA (post-hoc Bonferroni), and by Kruskal–
counts of these species were calculated. Wallis ANOVA with repeat Mann–Whitney U-tests.

Table 1 Bacterial species and subspecies included in the DNA–DNA checkerboard kit
Species panel 1 Collection Species panel 2 Collectiona

1a. Aggregatibacter actinomycetemcomitans (strain a) ATCC 29523 1. Actinomyces neuii GUH 550898
1b. Aggregatibacter actinomycetemcomitans (Y4) ATCC 43718 2. Aerococcus christensenii GUH 070938
2. Actinomyces israelii ATCC 12102 3. Anaerococcus vaginalis GUH 290486
3. Actinomyces naeslundii (type I + II) ATCC 43146 4. Atopobium parvulum GUH 160323
4. Actinomyces odontolyticus ATCC 17929 5. Atopobium vaginae GUH 010535
5. Campylobacter gracilis ATCC 33236 6. Bacteroides ureolyticus GUH 080189
6. Campylobacter rectus ATCC 33238 7. Bifidobacterium biavati GUH 071026
7. Campylobacter showae ATCC 51146 8. Bifidobacterium bifidum GUH 070962
8. Capnocytophaga gingivalis ATCC 33612 9. Bifidobacterium breve GUH 080484
9. Capnocytophaga ochracea ATCC 33596 10. Bifidobacterium longum GUH 180689
10. Capnocytophaga sputigena ATCC 33612 11. Corynebacterium nigricans GUH 450453
11. Eikenella corrodens ATCC 23834 12. Corynebacterium aurimucosum pseudogenitalium GUH 071035
12. Eubacterium saburreum ATCC 33271 13. Dialister spp. GUH071045
13a. Fusobacterium nucleatum nucleatum ATCC 25586 14a. Enterococcus faecalis GUH170812
13b. Fusobacterium nucleatum polymorphum ATCC 10953 14b. Enterococcus faecalis ATCC 29212
13c. Fusobacterium nucleatum naviforme ATCC 49256 15. Escherichia coli GUH 070903
14. Fusobacterium periodonticum ATCC 33693 16. Gardnerella vaginalis GUH 080585
15. Lactobacillus acidophilus ATCC 11975 17. Haemophilus influenzae ATCC 49247
16. Leptotrichia buccalis ATCC 14201 18. Helicobacter pylori ATCC 43504
17. Parvimonas micra ATCC 19696 19. Lactobacillus crispatus GUH 160342
18. Neisseria mucosa ATCC 33270 20. Lactobacillus gasseri GUH 170856
19. Prevotella intermedia ATCC 25611 21. Lactobacillus iners GUH 160334
20. Prevotella melaninogenica ATCC 25845 22. Lactobacillus jensenii GUH 160339
21. Prevotella nigrescens ATCC 33563 23. Lactobacillus vaginalis GUH 0780928
22. Porphyromonas gingivalis ATCC 33277 24. Mobiluncus curtisii GUH 070927
23. Propionibacterium acnes (type I + II) ATCC11827/28 25. Mobiluncus mulieris GUH 070926
24. Selenomonasnoxia ATCC 43541 26. Peptoniphilus spp. GUH 550970
25. Staphylococcus aureus ATCC 25923 27. Porphyromonas endodontalis ATCC35406
26. Streptococcus anginosus ATCC 33397 28. Peptostreptococcus anaerobius GUH 160362
27. Streptococcus constellatus ATCC 27823 (M32b) 29. Prevotella bivia GUH 450429
28. Streptococcus gordonii ATCC 10558 30. Prevotella disiens GUH 190184
29. Streptococcus intermedius ATCC 27335 31. Prevotella mirabilis GUH 070918
30. Streptococcus mitis ATCC 49456 32. Pseudomonas aeruginosa ATCC 33467
31. Streptococcus oralis ATCC 35037 33a. Staphylococcus aureus (yellow) GUH 070921
32. Streptococcus sanguinis ATCC 10556 33b. Staphylococcus aureus (white) GUH 070922
33. Streptococcus mutans ATCC 25175 34. Staphylococcus epidermidis GUH 130381
34. Tannerella forsythia ATCC 43037 (338) 35. Staphylococcus haemolyticus GUH071047
35. Treponema denticola ATCC 35405 36. Streptococcus agalactiae GUH 230282
36. Treponema socranskii D40DR2 37. Varibaculum cambriense GUH 070917
37. Veillonella parvula ATCC 10790

ATCC, American Type Culture Collection; D, sample from Forsyth Institute (Boston, Massachusetts); GUH, Ghent University Hospital Collection (Ghent, Belgium).
a
Strain a bacteria evaluated.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
242 European Journal of Gastroenterology & Hepatology 2013, Vol 25 No 2

Table 2 Clinical characteristics of patients with Crohn’s disease, ulcerative colitis, and control individuals
Crohn’s disease Ulcerative colitis Controls

Variables Mean SD Mean SD Mean SD ANOVA

Age 39.5 10.5 45.0 9.3 42.1 7.8 0.10


Number of teeth 22.8 5.6 21.0 5.1 23.8 4.2 0.19
Plaque index (%) 57.7 39.7 55.0 36.7 68.8 37.4 0.79
BOP (%) 37.1 – 29.5 – 31.5 – 0.84
Number of years with diagnosis of IBD 98.5 101.0 94.4 64.2 – – –
Smoking habit (%) – – – – – – 0.06
Smokers 21.4 – 6.7 – 6.7 – –
Nonsmokers 50.0 – 46.7 – 80.0 – –
Former smokers 28.6 – 46.7 – 13.3 – –

ANOVA, analysis of variance; BOP, bleeding on probing; IBD, inflammatory bowel disease.

The data were analyzed using a statistical software (IBM Discussion


SPSS 18.0; SPSS Inc., Armonk, New York, USA). Our study showed that the concentrations of B. ureolyticus,
C. gracilis P. micra, P. melaninogenica, P. anaerobius, S. aureus,
Results S. anginosus, S. intermedius, S. mitis, S. mutans, and T. denticola
Clinical data differed between patients with CH, ulcerative colitis, and
The demographic data from CD, UC, and control controls irrespective of the degree of periodontal
patients are reported in Table 2. Multiple-comparison destruction in inflamed sites. Patients with CD harbored
analysis showed that age, number of teeth, plaque index, the highest concentration of these bacteria. For the other
BOP, time of diagnosis of IBD, and smoking habit did not analyzed bacteria, no significant variation was observed
differ significantly between the groups. Multiple-compar- between the groups. The implication of these findings is
ison analysis did not show a significant difference unknown, but it is evident that inflamed periodontal
between the groups in any of the clinical parameters: regions can harbor harmful bacteria that can be impli-
PPD, CAL, and percentage of sites with plaque between cated not only in periodontal diseases but also in caries,
the groups either in gingivitis or in periodontitis sites. and most importantly, opportunistic infections.

Microbiological results In terms of bacteria involved with dental caries, CD


Gingivitis sites patients had higher counts of S. mutans than controls and
Multiple-comparison analysis showed that Parvimonas UC patients. Our finding of S. mutans in subgingival
micra, Prevotella melaninogenica, Peptostreptococcus anaerobius, plaque is in agreement with other studies [20–22]. It has
Staphylococcus aureus, Streptococcus anginosus, Streptococcus been shown previously that Streptococcus spp. is predomi-
mitis, S. mutans, and Treponema denticola differed signifi- nant in the inflamed intestinal mucosa of 80% of all
cantly between the groups (P < 0.001). The independent bacteria in CD patients, but not in UC patients [23].
analyses showed that all these bacteria were significantly Although S. mutans is a major pathogen in dental caries,
higher in CD patients compared with UC patients. this could to some extent explain the increased risk for
P. micra, P. melaninogenica, P. anaerobius, S. aureus, S. anginosus, dental caries that has been reported earlier in CD
and S. mutans were also significantly higher in CD patients [24–26].
patients compared with controls. When UC patients We also found that patients with CD had higher counts
were compared with controls, P. anaerobius and S. aureus of S. aureus than controls and UC patients. S. aureus is
were significantly higher, whereas P. micra, S. anginosus, commonly isolated from community and hospital infec-
S. mitis, and S. aureus were significantly lower than those tions affecting the lower respiratory tract, urinary tract,
of the control group (Table 3). skin, and bloodstream [27]. More than 70% of the
staphylococci isolated from bloodstream infections in the
Periodontitis sites ICU are of the species S. aureus, Staphylococcus epidermidis,
Multiple-comparison analysis showed that Bacteroides and Staphylococcus haemolyticus [28,29]. A high detection
ureolyticus, Campylobacter gracilis, P. melaninogenica, S. aureus, level of S. aureus has also been identified in saliva from
S. anginosus, Streptococcus intermedius, S. mitis, and S. mutans hospitalized patients [30]. Besides, S. aureus has also been
differed significantly between the groups (P < 0.001). identified in periodontitis [21,31,32]. Therefore, elim-
The two independent analyses showed that all these ination of inflamed sites in CD patients is not only
bacteria were significantly higher in CD when compared a measure to improve oral health but could also be
with UC patients. Except for S. mitis, they were also an important means to avoid nosocomial infections.
significantly higher in CD patients compared with
controls. UC and controls did not differ significantly Similarly, CD patients had significantly higher counts of
(Table 3). S. anginosus in both gingivitis and periodontitis sites when

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Subgingival flora in IBD patients Brito et al. 243

Table 3 Distributions of bacteria with differences in bacterial counts and significance levels, Crohn disease, ulcerative colitis, and control
groups in sites with gingivitis or periodontitis
Bacteria Patient group 25th 50th 75th Comparisons Significance

Gingivitis
Parvimonas micra Control 0.00 0.87 1.12 CD vs. UC 0.001
CD 0.98 1.36 2.53 CD vs. control 0.01
UC 0.00 0.47 0.85 UC vs. control 0.01
Prevotella melaninogenica Control 0.14 0.29 0.79 CD vs. UC 0.01
CD 0.80 1.26 2.01 CD vs. control 0.001
UC 0.22 0.30 0.89 UC vs. Control NS
Peptostreptococcus anaerobius Control 0.15 0.26 0.43 CD vs. UC 0.05
CD 0.31 0.62 1.04 CD vs. control 0.05
UC 0.14 0.21 0.54 UC vs. control 0.05
Staphylococcus aureus Control 0.28 0.36 0.71 CD vs. UC 0.01
CD 0.82 1.76 2.40 CD vs. control 0.001
UC 0.22 0.46 1.55 UC vs. control 0.01
Streptococcus anginosus Control 0.14 0.29 0.43 CD vs. UC 0.01
CD 0.53 0.97 1.82 CD vs. control 0.001
UC 0.12 0.19 0.41 UC vs. control 0.01
Streptococcus mitis Control 0.14 0.40 0.63 CD vs. UC 0.01
CD 0.24 0.74 1.25 CD vs. control NS
UC 0.10 0.21 0.25 UC vs. control 0.01
Streptococcus mutans Control 0.00 0.29 0.57 CD vs. UC 0.023
CD 0.00 0.78 1.66 CD vs. control 0.039
UC 0.00 0.18 0.33 UC vs. control NS
Treponema denticola Control 0.00 0.15 0.44 CD vs. UC 0.013
CD 0.20 0.54 1.39 CD vs. control NS
UC 0.00 0.17 0.22 UC vs. control NS
Periodontitis
Bacteroides ureolyticus Control 0.12 0.18 0.28 CD vs. UC 0.03
CD 0.30 0.48 0.62 CD vs. control 0.02
UC 0.14 0.21 0.28 UC vs. control NS
Campylobacter gracilis Control 0.14 0.38 0.71 CD vs. UC 0.001
CD 0.55 0.82 1.29 CD vs. control 0.05
UC 0.19 0.37 0.71 UC vs. control NS
P. melaninogenica Control 0.27 0.48 0.93 CD vs. UC 0.001
CD 0.74 1.34 2.32 CD vs. control 0.01
UC 0.25 0.46 0.69 UC vs. control NS
S. aureus Control 0.18 0.51 1.05 CD vs. UC 0.001
CD 0.78 1.34 2.33 CD vs. control 0.05
UC 0.28 0.47 0.63 UC vs. control NS
S. anginosus Control 0.14 0.32 0.54 CD vs. UC 0.001
CD 0.74 1.09 1.75 CD vs. control 0.001
UC 0.15 0.25 0.43 UC vs. control NS
S. intermedius Control 0.21 0.35 0.55 CD vs. UC 0.001
CD 0.55 0.78 1.25 CD vs. control 0.01
UC 0.20 0.34 0.49 UC vs. control NS
S. mitis Control 0.24 0.32 0.68 CD vs. UC 0.001
CD 0.44 0.66 1.00 CD vs. control NS
UC 0.12 0.29 0.41 UC vs. control NS
S. mutans Control 0.00 0.32 0.66 CD vs. UC 0.001
CD 0.34 0.86 1.49 CD vs. control 0.006
UC 0.19 0.22 0.48 UC vs. control NS

Numbers indicate  105 bacteria.


CD, Crohn’s disease; UC, ulcerative colitis.

compared with controls and UC patients. As S. anginosus is ontitis. Specifically, N. mucosa and Prevotella intermedia
an opportunistic bacterium that has been associated with were included among the 10 species in the control group
endocarditis [33–34], the diagnosis of periodontal dis- but not in CD or UC patients. Both S. aureus and
eases and consequently the elimination of inflamed sites S. haemolyticus were included in CD patients, and
through periodontal treatment is an important approach S. haemolyticus was found among the four highest-ranking
to eradicate an important reservoir of opportunistic species both in CD and in UC patients but were not
bacteria in IBD patients. included in the HC patients. Moreover, Firmicutes,
Bacteroidetes, and Proteobacteria identified as prevalent
Although the distribution of the 10 most dominating species in CD [35] were found in subgingival samples
species in CD, UC, and control individuals included more from our patients with CD and untreated periodontitis.
or less the same species, the ranking order differed. Large This may indicate a disturbed microbiota in CD patients
differences in bacterial counts were not expected because even presenting periodontal characteristics similar to
of the fact that the participants had untreated period- those of UC patients and controls.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
244 European Journal of Gastroenterology & Hepatology 2013, Vol 25 No 2

T. denticola, a motile microorganism, is recognized as a key 4 D’Aiuto F, Graziani F, Tetè S, Gabriele M, Tonetti MS. Periodontitis: from local
pathogen in periodontitis. It has been shown that infection to systemic diseases. Int J Immunopathol Pharmacol 2005; 18
(Suppl 3):1–11.
colonization by T. denticola above certain thresholds 5 Williams RC, Offenbacher S. Periodontal medicine: the emergence of a new
increases significantly the risk of periodontal disease branch of periodontology. Periodontol 2000 2000; 23:9–12.
progression in Chinese patients [36]. Besides, Byrne 6 Flemmig TF, Shanahan F, Miyasaki KT. Prevalence and severity of periodontal
disease in patients with inflammatory bowel disease. J Clin Periodontol
et al. [37] have reported that the proportions of 1991; 18:690–697.
Porphyromonas gingivalis and T. denticola in subgingival 7 Grössner-Schreiber B, Fetter T, Hedderich J, Kocher T, Schreiber S, Jepsen
plaque have the potential to help in the identification of S. Prevalence of dental caries and periodontal disease in patients with
inflammatory bowel disease: a case–control study. J Clin Periodontol 2006;
sites at significant risk for periodontitis progression. 33:478–484.
It can be speculated that CD patients have a higher risk 8 Brito F, de Barros FC, Zaltman C, Carvalho AT, Carneiro AJ, Fischer RG,
of disease progression than UC. The difference was found et al. Prevalence of periodontitis and DMFT index in patients with Crohn’s
disease and ulcerative colitis. J Clin Periodontol 2008; 35:555–560.
only in gingivitis sites, which indicated that major 9 Paster BJ, Boches SK, Galvin JL, Ericson RE, Lau CN, Levanos VA, et al.
differences may appear before CAL. This hypothesis Bacterial diversity in human subgingival plaque. J Bacteriol 2001;
183:3770–3783.
should be tested in a longitudinal study.
10 Gendron R, Grenier D, Maheu-Robert L. The oral cavity as a reservoir of
bacterial pathogens for focal infections. Microbes Infect 2000; 2:897–906.
It is unknown how various medications influence the
11 Viget N, Vernier-Massouille G, Salmon-Ceron D, Yazdanpanah Y, Colombel
growth of certain oral bacterial species. This may be JF. Opportunistic infections in patients with inflammatory bowel disease:
specifically important in patients who are medicated with prevention and diagnosis. Gut 2008; 57:549–558.
12 van Dyke TE, Dowell VR Jr, Offenbacher S, Snyder W, Hersh T. Potential
drugs that modulate the host inflammatory responses and role of microorganisms isolated from periodontal lesions in the pathogenesis
specifically as to how such medications may upregulate or of inflammatory bowel disease. Infect Immun 1986; 53:671–677.
downregulate proinflammatory cytokines. This should be 13 Sundh B, Johansson I, Emilson CG, Nordgren S, Birkhed D. Salivary
antimicrobial proteins in patients with Crohn’s disease. Oral Surg Oral Med
investigated more carefully, although we are aware that it is Oral Pathol 1993; 76:564–569.
a difficult task to group IBD patients according to the same 14 Docktor MJ, Paster BJ, Abramowicz S, Ingram J, Wang YE, Correll M, et al.
oral condition and also according to the medications they Alterations in diversity of the oral microbiome in pediatric inflammatory bowel
disease. Inflamm Bowel Dis 2012; 18:935–942.
are taking. IBD treatment is individual and the medications 15 Best WR, Becktel JM, Singleton JW, Kern F Jr. Development of a Crohn’s
used can be altered any time according to the patient’s disease activity index. National Cooperative Crohn’s Disease Study.
needs [38]. Besides the medication, the composition of the Gastroenterology 1976; 70:439–444.
16 Truelove SC, Witts LJ. Cortisone in ulcerative colitis; final report on a
intestinal microflora might also be influenced by different therapeutic trial. Br Med J 1955; 2:1041–1048.
dietary habits. De Filippo et al. [39] have examined the 17 Lindhe J, Ranney R, Lamster I, Charles A, Chung C-P, Flemmig T, et al.
human intestinal microbiota from children exposed to Consensus reports: chronic periodontitis. American Academy of
Periodontology. Ann Periodontol 1999; 4:38.
a modern Western diet and a rural diet, and reported that it 18 Socransky SS, Haffajee AD, Smith C, Martin L, Haffajee JA, Uzel NG, et al.
is important to preserve the microbial diversity from Use of checkerboard DNA-DNA hybridization to study complex microbial
ancient rural communities worldwide. We believe that ecosystems. Oral Microbiol Immunol 2004; 19:352–3562.
19 Persson GR, Weibel M, Hirschi R, Katsoulis J. Similarities in the subgingival
the patients analyzed in the present study were on a quite microbiota assessed by a curet sampling method at sites with chronic
similar diet; however, this information should be considered periodontitis. J Periodontol 2008; 79:2290–2296.
in further investigations. 20 Van der Reijden WA, Dellemijn-Kippuw N, Stijne-van Nes AM, de Soet JJ,
van Viget N, Vernier-Massouille G, et al. Opportunistic infections in patients
with inflammatory bowel disease: prevention and diagnosis. Gut 2008;
Conclusion 57:549–558.
This study showed that even presenting similar clinical 21 Fritschi BZ, Albert-Kiszely A, Persson GR. Staphylococcus aureus and other
bacteria in untreated periodontitis. J Dent Res 2008; 87:589–593.
periodontal parameters, IBD patients harbor higher levels 22 López R, Dahlén G, Retamales C, Baelum V. Clustering of subgingival
of bacteria that are related to opportunistic infections in microbial species in adolescents with periodontitis. Eur J Oral Sci 2011;
inflamed subgingival sites that might be harmful for the 119:141–150.
23 Fyderek K, Strus M, Kowalska-Duplaga K, Gosiewski T, Wedrychowicz A,
crucial microbe–host interaction. Jedynak-Wasowicz U, et al. Mucosal bacterial microflora and mucus layer
thickness in adolescents with inflammatory bowel disease. World J
Gastroenterol 2009; 15:5287–5294.
Acknowledgements 24 Schütz T, Drude C, Paulisch E, Lange KP, Lochs H. Sugar intake, taste
This study was supported in part by a grant from FAPERJ changes and dental health in Crohn’s disease. Dig Dis 2003; 21:252–257.
(Grant Number E26/101.528/2010). 25 Sundh B, Emilson CG. Salivary and microbial conditions and dental health in
patients with Crohn’s disease: a 3-year study. Oral Surg Oral Med Oral
Pathol 1989; 67:286–290.
26 Bevenius J. Caries risk in patients with Crohn’s disease: a pilot study.
Conflicts of interest Oral Surg Oral Med Oral Pathol 1988; 65:304–307.
There are no conflicts of interest. 27 Feng Y, Chen CJ, Su LH, Hu S, Yu J, Chiu CH. Evolution and pathogenesis
of Staphylococcus aureus: lessons learned from genotyping and
comparative genomics. FEMS Microbiol Rev 2008; 32:23–37.
28 Sader HS, Gales AC, Pfaller MA, Mendes RE, Zoccoli C, Barth A, et al.
References Pathogen frequency and resistance patterns in Brazilian hospitals: summary
1 Mariotti A. Dental plaque-induced gingival diseases. Ann Periodontol 1999; of results from three years of the SENTRY Antimicrobial Surveillance
4:7–19. Program. Braz J Infect Dis 2001; 5:200–214.
2 Flemmig TF. Periodontitis. Ann Periodontol 1999; 4:32–38. 29 Pereira EM, Schuenck RP, Malvar KL, Iorio NL, Matos PD, Olendzki AN,
3 Kinane DF, Mark Bartold P. Clinical relevance of the host responses of et al. Staphylococcus aureus, Staphylococcus epidermidis and
periodontitis. Periodontol 2000 2007; 43:278–293. Staphylococcus haemolyticus: methicillin-resistant isolates are detected

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Subgingival flora in IBD patients Brito et al. 245

directly in blood cultures by multiplex PCR. Microbiol Res 2010; 35 Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR.
165:243–249. Molecular-phylogenetic characterization of microbial community imbalances
30 Zuanazzi D, Souto R, Mattos MB, Zuanazzi MR, Tura BR, Sansone C, et al. in human inflammatory bowel diseases. Proc Natl Acad Sci USA 2007;
Prevalence of potential bacterial respiratory pathogens in the oral cavity of 104:13780–13785.
hospitalised individuals. Arch Oral Biol 2010; 55:21–28. 36 Papapanou PN, Baelum V, Luan WM, Madianos PN, Chen X,
31 Cuesta AI, Jewtuchowicz V, Brusca MI, Nastri ML, Rosa AC. Prevalence of Fejerskov O, et al. Subgingival microbiota in adult Chinese: prevalence
Staphylococcus spp and Candida spp in the oral cavity and periodontal and relation to periodontal disease progression. J Periodontol 1997;
pockets of periodontal disease patients. Acta Odontol Latinoam 2010; 68:651–666.
23:20–26. 37 Byrne SJ, Dashper SG, Darby IB, Adams GG, Hoffmann B, Reynolds EC.
32 Da Silva-Boghossian CM, do Souto RM, Luiz RR, Colombo AP. Association Progression of chronic periodontitis can be predicted by the levels of
of red complex, A. actinomycetemcomitans and non-oral bacteria with Porphyromonas gingivalis and Treponema denticola in subgingival plaque.
periodontal diseases. Arch Oral Biol 2011; 56:899–906. Oral Microbiol Immunol 2009; 24:469–477.
33 Woo PC, Tse H, Chan KM, Lau SK, Fung AM, Yip KT, et al. ‘Streptococcus 38 Kozuch PL, Hanauer SB. Treatment of inflammatory bowel disease:
milleri’ endocarditis caused by Streptococcus anginosus. Diagn Microbiol a review of medical therapy. World J Gastroenterol 2008; 14:
Infect Dis 2004; 48:81–88. 354–377.
34 Abdul-Redha RJ, Kemp M, Bangsborg JM, Arpi M, Christensen JJ. Infective 39 De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S,
endocarditis: identification of catalase-negative, Gram-positive cocci from et al. Impact of diet in shaping gut microbiota revealed by a comparative
blood cultures by partial 16S rRNA gene analysis and by Vitek 2 study in children from Europe and rural Africa. Proc Natl Acad Sci USA
examination. Open Microbiol J 2010; 4:116–122. 2010; 107:14691–14696.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

Вам также может понравиться