Topic 01

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

journal of dentistry 41 (2013) 779–786

Available online at www.sciencedirect.com

journal homepage: www.intl.elsevierhealth.com/journals/jden

Antimicrobial resistance and virulence traits of


Enterococcus faecalis from primary endodontic
infections

Renata Ximenes Lins a,*, Aurimar de Oliveira Andrade a,


Raphael Hirata Junior b, Melanie J. Wilson c, Michael A.O. Lewis c,
David W. Williams c, Rivail Antonio Sergio Fidel a
a
School of Dentistry, Rio de Janeiro State University, Rio de Janeiro, Brazil
b
School of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro, Brazil
c
School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, Wales, United Kingdom

article info abstract

Article history: Objectives: To determine the phenotypic and molecular characteristics of Enterococcus fae-
Received 10 May 2013 calis recovered from primary endodontic infections in Brazilian patients.
Received in revised form Methods: Twenty isolates of E. faecalis recovered from 43 Brazilian patients with primary
21 June 2013 endodontic infections were identified by biochemical profiling (API20Strep) and 16S rDNA
Accepted 2 July 2013 sequencing. Antimicrobial susceptibility was ascertained by agar dilution, using the recom-
mended protocol of the Clinical and Laboratory Standards Institute (CLSI). PCR with
validated primers was used to detect genes associated with antibiotic resistance and specific
Keywords: virulence factors.
Enterococcus faecalis Results: All isolates were deemed susceptible to penicillin G, erythromycin and vancomycin.
Genotype However, nine isolates had a minimum inhibitory concentration of 4 mg/mL to vancomycin
Antimicrobial susceptibility (the resistance breakpoint). Fourteen isolates (70% of isolates) were also resistant to
Endodontic infection tetracycline with MICs of >64 mg/mL. PCR products for tetracycline resistance genes were
detected in test isolates, while erythromycin and vancomycin resistance genes were not
evident. Gelatinase, aggregation substance and enteroccocal surface protein genes were
detected in 20, 18 and 12 isolates, respectively.
Conclusions: Endodontic E. faecalis isolates exhibit high level of resistance to tetracycline, an
antibiotic that has use in local treatment of dental infections. This opens up a much-needed
debate on the role and efficacy of this antibiotic for oral infections. Furthermore, these
isolates were shown to possess genes that could contribute to pathogenicity in the pulp
cavity.
# 2013 Elsevier Ltd. All rights reserved.

aetiological agents remain unclear. Ozok et al.1 suggested that


1. Introduction endodontic infections were more complex than previously
reported with more than 600 bacterial taxa associated with
The microbiome of primary and post treatment endodontic infected root canals. Although a single aetiological agent is
infections has been extensively studied, although the main unlikely, there is evidence supporting the association of

* Corresponding author at: Av Afranio de Mello Franco, 141/207 Leblon, CEP 22430-060, Rio de Janeiro, RJ, Brazil. Tel.: +55 21 2294 8268.
E-mail addresses: ximenes@uerj.br, endodontia@renataximenes.com.br (R.X. Lins).
0300-5712/$ – see front matter # 2013 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.jdent.2013.07.004
780 journal of dentistry 41 (2013) 779–786

enterococci, particularly Enterococcus faecalis with both primary The aims of this study were therefore to analyse isolates of
and secondary endodontic infection.2–8 E. faecalis from patients with primary endodontic infections for
Enterococci are Gram-positive, coccus-shaped bacteria and the presence of defined virulence genes and susceptibility to
frequent colonisers of the human gastrointestinal and antibiotics commonly used in dentistry and other areas of
genitourinary tracts. In the past, these bacteria were not clinical importance. Through enhanced awareness of the
considered particularly virulent. However, recent years have characteristics of E. faecalis involved in endodontic infection,
seen an increase in nosocomial infections caused by entero- we can identify optimal treatment strategies to improve
coccal species largely attributed to their antimicrobial resis- patients’ health and potentially limit dissemination of
tance profiles.9 Of the group, E. faecalis is considered the most antimicrobial resistance.
significant species, both in terms of frequency of isolation
from sites of infection and in the transfer of antibiotic
resistance.10 2. Materials and methods
Enterococci are regarded as antibiotic-resistant, opportu-
nistic pathogens frequently recovered from patients who have 2.1. Patient selection and sample collection
received multiple courses of antibiotics and who have been
hospitalized for prolonged periods. These bacteria exhibit A total of 43 patients with primary endodontic infection were
intrinsic resistance to several antibiotics, as well as an ability recruited with informed consent for participation in this
to rapidly acquire antibiotic resistance. Enterococci are not study. Only necrotic pulps without oral exposition were
generally regarded as normal inhabitants of the oral cavity, included. All patients were otherwise healthy and had not
but may colonise transiently, particularly in debilitated been hospitalised or had received antibiotics for at least 6
individuals.11 E. faecalis has, however, been isolated from a months prior to the study. Pregnancy or active periodontal
range of oral conditions including carious lesions, chronic disease were additional exclusion criteria. The research was
periodontitis, and has been associated with persistent apical approved by the Research Ethics Committee of Rio de Janeiro
periodontitis.12 E. faecalis has been detected primarily at sites State University (COEP051/2009).
of persistent endodontic infections,9,13–15 and has also been After plaque removal and rubber dam isolation, the tooth
found in high numbers in some primary endodontic infec- and the operative field was cleansed with 3% (v/v) hydrogen
tions.7,8,16 peroxide and then disinfected with 2.5% NaOCl solution.
Neither the source, nor the role of enterococci in the Coronal access was made using sterile round burs without
pathogenesis of endodontic infections is understood.15,17 Oral water spray. Samples were collected from the root canal using
E. faecalis has been shown to possess a range of virulence a #15 H-type file (Dentsply/Maillefer, Ballaigues, Switzerland)
factors including gelatinase activity, expression of haemoly- and two sterile paper points, as previously described.24 Both
sin, response to pheromones, bacteriocin production, biofilm file and paper points were then transferred to tubes containing
development, expression of adherence factors, aggregation Enterococcosel broth (Becton Dickinson Microbiology Sys-
substances and resistance to several antibiotics.12,18–21 Impor- tems, MD, USA) and incubated for 48 h at 37 8C to selectively
tantly, enterococci have been shown to transfer certain recover enterococci. Isolated colonies were then subcultured
virulence traits to related species in root canals ex vivo, on to blood agar plates (Becton Dickinson Microbiology
highlighting the need to more fully understand the role of Systems) and pure colonies subjected to identification tests.
these pathogens in infection in general.22 A sterile paper point without previous contact with the root
Finally, it is not unrealistic to expect that enterococci canal was included in all sampling procedures and served as a
resistant to multiple antimicrobials may be able to colonise negative control.
the root canal environment. Although Vancomycin Resistant
Enterococci (VRE) have not been isolated in root canal 2.2. Culture and identification of E. faecalis
infections, Nandakumar et al.23 identified proteins from the
tet and van operons in all the patient cases where enterococci All isolates were presumptively identified as enterococci based
were detected by genomic analysis of endodontic infections. on growth in the presence of bile and azide, and esculin
Additionally, in the patient sample from which the VanE hydrolysis. Additional tests to further characterise these
protein was identified, E. faecalis was highly prevalent. This is isolates included Gram-staining, catalase activity, motility
important from a treatment perspective because antibiotics testing, pyruvate fermentation, and an ability to grow in Tryptic
are known not to be very effective in treating chronic or Soy Agar (Merk Darmstadt, DE) supplemented with 6.5% NaCl at
localised acute endodontic infections or in preventing 42 8C. A commercial biochemical profiling test (API 20 Strep
recurrent episodes of infection. Besides, it is an alert to the identification kit; Analytical Profile Index; bioMérieux SA,
potential increase in antibiotic resistance amongst oral Marcy-Etoile, France) was also used, and E. faecalis ATCC
isolates. 29212 served as a positive control. The prevalence of E. faecalis
All this information justifies the isolation, identification was recorded as the percentage of cases examined.
and characterisation of virulence factors and antimicrobials
resistant determinants of endodontic enterococci. Genetic 2.3. Confirmation of E. faecalis identity by sequencing of
analysis can be used as an initial screening tool, and although 16S ribosomal DNA
detection of the target gene itself does not necessarily mean
that the encoded protein will subsequently be expressed, it Total DNA was extracted using the PureLinkTM Genomic DNA
does indicate the potential for expression. Mini Kit (Invitrogen, Paisley, UK) and the manufacturer’s
journal of dentistry 41 (2013) 779–786 781

recommended protocol. Extracted DNA served as template identity to sequences deposited in the public domain
for PCR using the universal bacterial 16S rRNA primer databases were considered to be positive identification of
pair of 27f and 1492r (Table 1). All PCRs were performed in a taxa.
50 mL final reaction volume containing 1 mM of each primer,
25 mL of PCR Master Mix (Promega, Madison, USA) and 2 mL 2.4. Antimicrobial susceptibility testing
of total DNA template. Negative controls of sterile ultrapure
water in place of DNA template were included with each The minimum inhibitory concentration (MIC) of E. faecalis
PCR. isolates against a range of antibiotics was determined by agar
PCR was undertaken in a DNA thermocycler using an dilution, using the recommendations of the Clinical and
initial denaturation step of 95 8C for 1 min, and then 26 Laboratory Standards Institute (CLSI). Briefly, Müeller-Hinton
thermal cycles of 94 8C for 45 s, 50 8C for 45 s and 72 8C for Agar (Difco, USA) plates were prepared with penicillin,
1.5 min, followed by a final step of 72 8C for 15 min. Gel erythromycin, tetracycline and vancomycin microdilutions
electrophoresis of the amplicons was performed in a 1.0% and inoculated with a bacterial suspension equivalent to 0.5
agarose gel at 60 V/cm2. PCR amplicons stained with safe McFarland standard and incubated at 37 8C for 18 h. Isolates
view dye (NBS Biologicals, Cambridgeshire, UK) were were considered susceptible or resistant according to break-
subsequently detected by ultraviolet transillumination. A point values recommended by the CLSI document M100-S21
100-bp DNA ladder (Promega) served as a molecular weight (Wayne, PA, USA).
standard.
Enterococcus species were then identified using Eurofins 2.5. Detection of antimicrobial resistance and virulence
MWG Operon’s sequencing service. Briefly, PCR products genes
were purified using the QIAquick PCR purification kit (Qiagen,
Hilden, Germany) and the DNA subjected to a sequencing PCR The DNA of all isolates was analysed for the presence of
reaction using the primer 357f (Table 1). The partial 16S rDNA antibiotic resistance genes including those for erythromycin,
sequences obtained were then compared with those tetracycline and vancomycin, and also for the virulence
sequences within the NCBI database using the Basic Local associated genes of gelatinase, surface protein, cytolysin
Alignment Search Tool (BLAST). Sequences with 98–100% and aggregation substance (Table 1).

Table 1 – PCR details to detect antimicrobial resistance to erythromycin (erm(A) and erm(B)), tetracycline (tet(M) and tet(L)),
vancomycin (vanA, vanB, vanC1, vanC2/3) and virulence-associated genes gelatinase (gel(E)), enterococcal surface protein
(esp), cytolysin (cylB), and aggregation substance (agg, asa373).
Gene target(s) Primer Oligonucleotide sequence (50 –30 ) Product size (bp) Tm (8C) Reference
26,28
16S RNA 27f GTGCTGCAGAGAGTTTGATCCTGGCTCAG 1164 54
1492r CACGGATCCTACGGGTACCTTGTTACGACTT
357f CTCCTACGGGCGGCAGCAG
29
erm(A) ERMA1 TCTAAAAAGCATGTAAAAGAA 645 50
ERMA2 CTTCGATAGTTTATTAATATTAGT
29
erm(B) ERMB1 GAAAAGGTACTCAACCAAATA 639 52
ERMB2 AGTAACGGTACTTAAATTGTTTC
27
tet(M) TETM1 AGTTTTAGCTCATGTTGATG 1862 55
TETM2 TCCGACTATTTAGACGACGG
27
tet(L) TETL1 CCTGCGAGTACAAACTGG 1209 55
TETL2 TCAAGGTAACCAGCCAAC
25
vanA VANA1 GGGAAAACGACAATTGC 732 50
VANA2 GTACAATGCGGCCGTTA
25
vanB VANB ATGGGAAGCCGATAGTC 635 52
VANB2 GATTTGCTTCCTCGACC
25
vanC1 VANC1-1 GGTATCAAGGAAACCTC 822 50
VANC1-2 CTTCCGCCATCATAGCT
25
vanC2/3 VANC2/3-1 CTCCTACGATTCTCTTG 439 50
VANC2/3-2 CGAGCAAGACCTTTAAG
25
gelE GELE1 ACGCATTGCTTTTCCATC 419 51
GELE2 ACCCCGTATCATTGGTTT
25
esp ESP1 TTGCTAATGCTAGTCCACGACC 932 58
ESP2 GCGTCAACACTTGCATTGCCA
25
cylB CYLB1 ATTCCTACCTATGTTCTGTTA 843 50
CYLB2 AATAAACTCTTCTTTTCCAAC
25
agg AGG1 AAGAAAAAGAAGTAGACCAAC 1555 52
AGG2 AAACGGCAAGACAAGTAAATA
25
asa373 ASA373F GGACGCACGTACACAAAGCTACC 619 58
ASA373R TGGGTGTGATTCCGCTGTA
782 journal of dentistry 41 (2013) 779–786

and the possible high recovery based on use of an Enterococcus


3. Results selective isolation medium. The observed high prevalence,
does however, highlight the importance of continuous
3.1. E. faecalis isolates surveillance of E. faecalis at various sampling sites and the
impact of population demographics that could influence
A total of 20 E. faecalis isolates were recovered from the 43 microbial communities such as these.
patients studied following use of the selective medium for The origin and role of enterococci in the pathogenesis of
Enterococcus. endodontic infection is also unclear.7,12,17,34–36 Regarding the
role of enterococci in endodontic infection, we believe that
3.2. Antibiotic susceptibility knowledge of bacterial virulence factors involved in the
pathogenesis of apical periodontitis is fundamental to
All isolates were deemed susceptible to penicillin G and understanding the disease process and to assist in establish-
erythromycin, being inhibited by 2 and 0.25 mg/mL, respec- ing appropriate treatment, motivating us to characterise the
tively (Table 2).30 All isolates were susceptible to vancomycin, endodontic isolates both genotypically and phenotypically.
with MICs raging from 1 to 4 mg/mL. However, 9 of these In the present study, antimicrobial susceptibility was
‘susceptible’ isolates did exhibit MICs of 4 mg/mL to vancomy- determined for all endodontic isolates, and revealed a high
cin, and this is the resistance breakpoint concentration. An prevalence of tetracycline resistance (70%), exceeding
MIC above 4 mg/mL, i.e. between 8 mg/mL and 16 mg/mL would previously reported findings.2,18,37 The incidence does however,
result in isolates being classed as having ‘intermediate appear to be increasing, for example, between 2000 and 2010,
resistance’ to vancomycin. Fourteen strains (Table 2) were several studies have reported E. faecalis from endodontic
resistant to tetracycline, with MICs >64 mg/mL, representing infections to exhibit resistance to tetracycline at levels
70% of the isolates. increasing from 14.3%, 15.1%, 28.8% and 30% according to
Pinheiro et al.,38 Sedgley et al.,19 Reynaud af Geijersstam et al.,39
3.3. Detection of antimicrobial resistance and virulence and Skucaite et al.,40 respectively.
genes Kuch et al.41 analysed antimicrobial susceptibility of 386 E.
faecalis isolates from hospitals and communities of six
PCR products for tetracycline resistance were detected in European countries. The percentage of resistance to tetracy-
several test isolates and the relative prevalence of these genes cline was 77.9% for hospital isolates and 55.6% for community
are presented in Table 3.30 The tetM gene was detected in 12 isolates. In our study, we have therefore encountered a higher
isolates, whilst tetL was present in 4 isolates, representing 60% prevalence of tetracycline in endodontic isolates compared to
and 20% of the isolates, respectively. The ermA, ermB, vanA, those previously noted for community isolates.
vanB, vanC1 and vanC2/3 genes were not detected in any tested
isolates (Table 3).
PCR for specific virulence genes (Table 3) showed that all 20
isolates (100%) were positive for gelatinase (gelE), whilst Table 2 – Antibiotic susceptibility characteristics of E.
faecalis. Susceptibility (S) and resistance (R) MIC break-
enteroccocal surface protein (esp) gene was detected for 13
points (mg/mL) as recommended by CLSI (2011): penicillin
isolates (65%), and aggregation substance (agg) gene was found
(=8 S, I16 R); erythromycin (=0.5 S, I8 R); tetracycline
in 18 isolates (90%). All isolates were negative for the asa373 (=4 S, I16 R) and vancomycin (=4 S, I32 R).
(aggregation substance) gene and cytolysin (cylB) gene.
Isolate MIC (mg/mL)
Penicillin G Erythromycin Tetracycline Vancomycin
4. Discussion 1 2(S) 0.25(S) >64(R) 4(S)
2 2(S) 0.25(S) >64(R) 4(S)
3 2(S) 0.25(S) 1(S) 2(S)
Despite the microbiome of primary and post treatment
4 2(S) 0.25(S) 16(S) 2(S)
endodontic infections being extensively studied, the main 5 2(S) 0.25(S) 4(S) 2(S)
aetiological agents remain unclear. There is evidence support- 6 2(S) 0.25(S) 4(S) 2(S)
ing an association of enterococci, particularly E. faecalis with 7 2(S) 0.25(S) >64(R) 2(S)
both primary and secondary endodontic infection.2–8 Reports 8 2(S) 0.25(S) >64(R) 4(S)
on E. faecalis prevalence in root canals can vary considerably, 9 2(S) 0.25(S) >64(R) 4(S)
10 2(S) 0.25(S) >64(R) 4(S)
possibly due to differences in methods of clinical sampling
11 2(S) 0.25(S) >64(R) 2(S)
and analysis. In teeth exhibiting failed endodontic treatment,
12 2(S) 0.25(S) >64(R) 4(S)
previous studies report an E. faecalis prevalence of between 13 2(S) 0.25(S) 4(S) 2(S)
18.5–70% when culture is used, and 67–89.6% using PCR 14 2(S) 0.25(S) 4(S) 1(S)
detection.5,7,14,16,31–33 However, in cases of primary endodontic 15 2(S) 0.25(S) >64(R) 4(S)
infection, prevalence is lower, ranging between 4–12.5% with 16 2(S) 0.25(S) >64(R) 4(S)
culture and 33–89.3% using PCR.7,8,14,16 In this study, a high 17 2(S) 0.25(S) >64(R) 4(S)
18 2(S) 0.25(S) >64(R) 2(S)
prevalence of E. faecalis was noted (20/43 patients), and the
19 2(S) 0.25(S) >64(R) 2(S)
exact reasons for this remain unclear. There are several 20 2(S) 0.25(S) >64(R) 2(S)
factors that could have been responsible for this high recovery
Total (%) 0 0 75 0
which may relate to the demographics of the patients studied
journal of dentistry 41 (2013) 779–786 783

Table 3 – Genetic profiles of E. faecalis from primary endodontic infection in Brazil.


Isolate Antimicrobial resistance genes Virulence genes
ermA ermB tet tet vanA vanB van van gelE esp agg asa cylB
M L C1 C2/3 373
1   +      + + +  
2         + + +  
3         + + +  
4         +  +  
5         +  +  
6         +  +  
7   + +     +  +  
8   +      + + +  
9   +      + + +  
10   +      + + +  
11         +  +  
12   +      + + +  
13         + +   
14         +    
15   +      + +   
16   +      + +   
17   +      + +   
18   + +     +  +  
19   + +     + + +  
20   + +     + + +  

Total (%) 0 0 60 20 0 0 0 0 100 65 90 0 0

This increased incidence of resistance to tetracycline for use of tetracycline in endodontic infection is still being utilised
endodontic isolates may be explained by selective pressure in some cases, and may contribute to an increase in resistance
due to previous use of tetracyclines in the practice of dentistry, to this antibiotic by exposed isolates.
such as in the treatment of localised aggressive periodontitis PCR products for tetracycline resistance genes were also
and as an intracanal medication. In a longitudinal study over detected in our study, with the tetM gene being evident in 12
12 months, Rodrigues et al.42 observed the antibiotic resis- isolates and tetL in 4 isolates, representing 60% and 20% of the
tance profile of subgingival microbiota following systemic and samples. There was a direct correlation of 85% between the
local administration of tetracycline as an adjunct to periodon- presence of the tetM and antibiotic susceptibility test result (12
tal therapy. In all cases where tetracycline was used, there was of 14 tetracycline resistant isolates were positive to tetM)
an initial selection of drug-resistant microorganisms, and only indicating that PCR analysis may be an important marker for
after months of continuous treatment, particularly topically screening antibiotic resistance. Villedieu et al.46 also found a
and at high concentrations was their prevalence reduced. This high prevalence of 79% for the gene tetM in oral bacteria
fact demonstrates that short-term use of such intracanal isolated from dental plaque and saliva.
medication may promote prevalence of resistant strains at the Tetracycline resistance in clinical isolates of enterococci is
site of infection. frequently associated with plasmids and/or conjugative
In endodontics, several studies have assessed the antimi- transposons47 which can readily be transferred between
crobial efficacy of antibiotic mixtures containing ciprofloxa- species.48
cin, metronidazole, and minocycline against the pathogens The conditions in root canals of low nutrient levels and the
commonly found inside the root canal system including E. presence of antimicrobials agents limit sensitive strains and
faecalis. This triple antibiotic mixture is also promoted as an select for more resistant species such as E. faecalis. Multiple
intracanal medicament to disinfect the root canal system to antibiotic resistance in clinical isolates of E. faecalis from root
encourage revascularization of a tooth with a necrotic pulp.43 canal infections has been reported,19,38,39,49 as well as the
Antibiotics have been suggested as possible irrigants by transfer of antibiotic resistance genes between these bacteria
Torabinejad et al.44 and Giardino et al.45 with both of these and related species.22,50 Although our study did not find multi-
studies including tetracycline within their irrigant formula- resistant isolates, or VRE, the possibility that these strains
tions. Tetracycline is also present in Ledermix paste (Haupt could occur in root canals infections cannot be excluded. In
Pharma GmbH, Wolfratshausen, Germany), a combination of our study, 9 isolates (45%) had MICs of 4 mg/mL to vancomycin
the same tetracycline antibiotic, demeclocycline HCl (3.2%), which is at the resistance breakpoint for this agent. This result
and a corticosteroid, triamcinolone acetonide (1%), in a motivated our investigation of the most common genes
polyethylene glycol base, which is advocated as an intracanal related to resistance to vancomycin in enterococci, namely,
dressing in cases of pulp-less infected root canals, pulp vanA, vanB, vanC1 and vanC2/3, which were not detected, thus
necrosis and infection with incomplete root formation confirming the MIC results of the present study and those
(apexification), perforations, inflammatory root resorption, previously reported.19,21,35,49 However, Nandakumar et al.23
inflammatory periapical bone resorption, and for the treat- found proteins from the tet and van operons to be predominant
ment of large periapical radiolucent lesions. Thus, the local in all patient cases where enterococci were detected by
784 journal of dentistry 41 (2013) 779–786

genomic analysis. ErmA and ermB genes were not detected in particularly in relation to the possession and expression of
our endodontic isolates, thus correlating with the phenotypic virulence factors in the root canal environment, to better
results of susceptibility in the MIC test. These findings were inform our management strategies. Environmental pressure
also in agreement with the work of Sedgley et al.19 in root canals may be responsible for the selection of more
There is ongoing debate regarding the relative merits of resistant species and the possession of virulence determi-
using local antibiotics to treat oral infections, particularly nants. This knowledge is important in guiding procedures for
given the inability of many antibiotics to penetrate bio- controlling the increasing problem of antibiotic resistance
films.51,52 New approaches have been proposed for biofilm amongst clinically relevant bacteria.
control by blocking adherence or co-aggregation mechanisms,
disruption of quorum-sensing mechanisms and probiotic
therapy.53 An understanding of the interactions between references
the microorganisms and which are the virulence factors
involved, provides opportunities for novel ‘non-antibiotic’
methods of control. In this context, we also investigated a 1. Ozok AR, Persoon IF, Huse SM, Keijser BJ, Wesselink PR,
number of virulence genes described in oral E. faecalis, Crielaard W, et al. Ecology of the microbiome of the infected
root canal system: a comparison between apical and coronal
including cytolysin (cylB), enterococcal surface protein (esp),
root segments. International Endodontic Journal 2012;45:
gelatinase (gelE) and aggregation substances (agg and asa373).
530–41.
Cytolysin is a haemolytic toxin that also has bacteriocin 2. Heintz CE, Deblinger R, Oliet S. Antibiotic sensitivities of
activity54; ESP is a surface protein that enhances biofilm enterococci isolated from treated root canals. Journal of
formation by E. faecalis,55 and gelatinase is a metalloprotease Endodontics 1975;1:373–6.
that cleaves several substrates including insoluble collagen 3. Möller AJ, Fabricius L, Dahlén G, Ohman AE, Heyden G.
fragments, as well as the pheromone and inhibitor peptides Influence on periapical tissues of indigenous oral bacteria
and necrotic pulp tissue in monkeys. Scandinavian Journal of
involved in conjugative plasmid transfer of E. faecalis. A role for
Dental Research 1981;89:475–84.
gelE in biofilm development has also been described.56 4. Sundqvist G. Ecology of the root canal flora. Journal of
Aggregation substance is a pheromone-inducible surface Endodontics 1992;18:427–30.
protein that promotes aggregation during bacterial conjuga- 5. Peciuliene V, Balciuniene I, Eriksen HM, Haapasalo M.
tion. This virulence trait and others factors, including Isolation of Enterococcus faecalis in previously root-filled
cytolysin and ESP were found to be encoded on a large, 153- canals in a Lithuanian population. Journal of Endodontics
2000;26:593–5.
kb pathogenicity island.57
6. Gomes BPFA, Pinheiro ET, Gade-Neto CR. Microbiological
In our study, the gelE was detected in all isolates, whilst esp
examination of infected dental root canals. Oral Microbiology
was detected in 13 isolates (65%), and agg was evident for 18 and Immunology 2004;19:71–6.
isolates (90%). As these virulence determinants have been 7. Sedgley C, Nagel A, Dahlén G, Reit C, Molander A. Real-time
shown to influence biofilm formation58 they may play a role in quantitative polymerase chain reaction and culture
promoting the persistence of enterococci in the root canal analyses of Enterococcus faecalis in root canals. Journal of
environment. Endodontics 2006;32:173–7.
8. Sassone L, Fidel R, Figueiredo L, Fidel S, Faveri M, Feres M.
Wang et al.20 analysed the relationship of biofilm formation
Evaluation of the microbiota of primary endodontic
and gelE gene expression in E. faecalis recovered from root
infections using checkerboard DNA–DNA hybridization. Oral
canals and found higher expression in cases of apical Microbiology and Immunology 2007;22:390–7.
radiolucency and in biofilm forming isolates. These authors 9. Bonten MJ, Willems R, Weinstein RA. Vancomycin-resistant
suggested that high expression of gelE may contribute to the enterococci: why are they here, and where do they come
development of apical periodontitis. Although gelatinase from. The Lancet Infectious Diseases 2001;1:314–25.
production in vitro was not examined in our study, other 10. Fisher K, Phillips C. The ecology, epidemiology and
virulence of Enterococcus. Microbiology 2009;155:1749–57.
studies have related expression to a determinant (ef1841/fsrC)
11. Murray BE. Vancomycin-resistant enterococcal infections.
of a deletion sequence of the fsr gene cluster, the quorum The New England Journal of Medicine 2000;342:710–21.
sensing system that regulate the expression of gelE gene. 12. Zhu X, Wang Q, Zhang C, Cheung GS, Shen Y. Prevalence,
Gelatinase-negative phenotype is related to the presence of phenotype, and genotype of Enterococcus faecalis isolated
this determinant.25 from saliva and root canals in patients with persistent
apical periodontitis. Journal of Endodontics 2010;36:1950–5.
13. Pinheiro ET, Gomes BPFA, Ferraz CCR, Teixeira FB, Zaia AA,
5. Conclusion Souza-Filho FJ. Evaluation of root canal microorganisms
isolated from teeth with endodontic failure and their
antimicrobial susceptibility. Oral Microbiology and
In summary, this study demonstrated a high incidence of Immunology 2003;18:100–3.
tetracycline resistance (an antibiotic that is still used in local 14. Rôças IN, Siqueira Jr JF, Santos KRN. Association of
treatment of oral infections) in E. faecalis isolates from primary Enterococcus faecalis with different forms of periradicular
endodontic infections. This opens up a much needed debate diseases. Journal of Endodontics 2004;30:315–20.
15. Kaufman B, Spangberg L, Barry J, Fouad AF. Enterococcus spp.
on the role and efficacy of this antibiotic for endodontic
in endodontically treated teeth with and without
infections. Furthermore, isolates were shown to possess genes
periradicular lesions. Journal of Endodontics 2005;31:851–6.
that could contribute to their pathogenicity in the pulp cavity, 16. Gomes BP, Pinheiro ET, Sousa EL, Jacinto RC, Zaia AA, Ferraz
and which can also be transferred in the microenvironment of CC, et al. Enterococcus faecalis in dental root canals detected
the root canal. Clearly, more research in this area is required by culture and by polymerase chain reaction analysis. Oral
journal of dentistry 41 (2013) 779–786 785

Surgery Oral Medicine Oral Pathology Oral Radiology and periradicular lesions by culture dependent and independent
Endodontic 2006;102:247–53. approaches. Journal of Endodontics 2006;32:722–6.
17. Zoletti GO, Pereira EM, Schuenck RP, Teixeira LM, Siqueira Jr 34. Vidana R, Sullivan A, Billström H, Ahlquist M, Lund B.
JF, dos Santos KR. Characterization of virulence factors and Enterococcus faecalis infection in root canals – host-derived or
clonal diversity of Enterococcus faecalis isolates from treated exogenous source? Letters in Applied Microbiology
dental root canals. Research in Microbiology 2011;162:151–8. 2011;52:109–15.
18. Dahlen G, Samuelsson W, Molander A. Identification and 35. Dahlén G, Blomqvist S, Almståhl A, Carlén A. Virulence
antimicrobial susceptibility of enterococci isolated from factors and antibiotic susceptibility in enterococci isolated
root canal. Oral Microbiology and Immunology 2000;15:309–12. from oral mucosal and deep infections. Journal of Oral
19. Sedgley CM, Molander A, Flannagan SE, Nagel AC, Appelbe Microbiology 2012;4:10855.
OK, Clewell DB, et al. Virulence, phenotype and genotype 36. Souto R, Colombo AP. Prevalence of Enterococcus faecalis in
characteristics of endodontic Enterococcus spp. Oral subgingival biofilm and saliva of subjects with chronic
Microbiology and Immunology 2005;20:10–9. periodontal infection. Archives of Oral Biology 2008;53:
20. Wang L, Dong M, Zheng J, Song Q, Yin W, Li J, et al. 155–60.
Relationship of biofilm formation and gelE gene expression 37. Noda M, Komatsu H, Inoue S, Sano H. Antibiotic
in Enterococcus faecalis recovered from root canals in patients susceptibility of bacteria detected from the root canal
requiring endodontic retreatment. Journal of Endodontics exudate of persistent apical periodontitis. Journal of
2011;37:631–6. Endodontics 2000;26:221–4.
21. Sun J, Sundsfjord A, Song X. Enterococcus faecalis from 38. Pinheiro ET, Gomes BP, Drucker DB, Zaia AA, Ferraz CC,
patients with chronic periodontitis: virulence and Souza-Filho FJ. Antimicrobial susceptibility of Enterococcus
antimicrobial resistance traits and determinants. European faecalis isolated from canals of root filled teeth with
Journal of Clinical Microbiology & Infectious Diseases periapical lesions. International Endodontic Journal
2012;31:267–72. 2004;37:756–63.
22. Sedgley CM, Lee EH, Martin MJ, Flannagan SE. Antibiotic 39. Reynaud af Geijersstam AH, Ellington MJ, Warner M,
resistance gene transfer between Streptococcus gordonii and Woodford N, Haapasalo M. Antimicrobial susceptibility and
Enterococcus faecalis in root canals of teeth ex vivo. Journal of molecular analysis of Enterococcus faecalis originating from
Endodontics 2008;34:570–4. endodontic infections in Finland and Lithuania. Oral
23. Nandakumar R, Madayiputhiya N, Fouad AF. Proteomic Microbiology and Immunology 2006;21:164–8.
analysis of endodontic infections by liquid 40. Skucaite N, Peciuliene V, Vitkauskiene A, Machiulskiene V.
chromatography–tandem mass spectrometry. Oral Susceptibility of endodontic pathogens to antibiotics in
Microbiology and Immunology 2009;24:347–52. patients with symptomatic apical periodontitis. Journal of
24. Sassone LM, Fidel RA, Faveri M, Guerra R, Figueiredo L, Fidel Endodontics 2010;36:611–6.
SR, et al. A microbiological profile of symptomatic teeth with 41. Kuch A, Willems RJ, Werner G, Coque TM, Hammerum AM,
primary endodontic infections. Journal of Endodontics Sundsfjord A, et al. Insight into antimicrobial susceptibility
2008;34:541–5. and population structure of contemporary human
25. Biavasco F, Foglia G, Paoletti C, Zandri G, Magi G, Enterococcus faecalis isolates from Europe. Journal of
Guaglianone E, et al. VanA-type enterococci from humans, Antimicrobial Chemotherapy 2012;67:551–8.
animals, and food: species distribution, population 42. Rodrigues RM, Gonçalves C, Souto R, Feres-Filho EJ, Uzeda
structure, Tn1546 typing and location, and virulence M, Colombo AP. Antibiotic resistance profile of the
determinants. Applied and Environmental Microbiology subgingival microbiota following systemic or local
2007;73:3307–19. tetracycline therapy. Journal of Clinical Periodontology
26. Frank JA, Reich CI, Sharma S, Weisbaum JS, Wilson BA, 2004;31:420–7.
Olsen GJ. Critical evaluation of two primers commonly used 43. Banchs F, Trope M. Revascularization of immature
for amplification of bacterial 16S rRNA genes. Applied and permanent teeth with apical periodontitis: new treatment
Environmental Microbiology 2008;74:2461–70. protocol. Journal of Endodontics 2004;30:196–200.
27. Schwaiger K, Harms K, Hölzel C, Meyer K, Karl M, Bauer J. 44. Torabinejad M, Khademi AA, Babagoli J, Cho Y, Johnson WB,
Tetracycline in liquid manure selects for co-occurrence of Bozhilov K, et al. A new solution for the removal of the
the resistance genes tet(M) and tet(L) in Enterococcus faecalis. smear layer. Journal of Endodontics 2003;29:170–5.
Veterinary Microbiology 2009;18:386–92. 45. Giardino L, Ambu E, Becce C, Rimondini L, Morra M. Surface
28. Turner S, Pryer KM, Miao VPW, Palmer JD. Investigating tension comparison of four common root canal irrigants
deep phylogenetic relationships among cyanobacteria and and two new irrigants containing antibiotic. Journal of
plastids by small subunit rRNA sequence analysis. Journal of Endodontics 2006;32:1091–3.
Eukaryotic Microbiology 1999;46:327–38. 46. Villedieu A, Diaz-Torres ML, Hunt N, McNab R, Spratt DA,
29. Zou LK, Wang HN, Zeng B, Li JN, Li XT, Zhang AY, et al. Wilson M, et al. Prevalence of tetracycline resistance genes
Erythromycin resistance and virulence genes in Enterococcus in oral bacteria. Antimicrobial Agents and Chemotherapy
faecalis from swine in China. New Microbiologica 2011;34: 2003;47:878–82.
73–80. 47. Clewell DB. Movable genetic elements and antibiotic
30. Hanningan A, Lynch C. Statistical methodology in oral and resistance in enterococci. European Journal of Clinical
dental research: pitfalls and recommendations. Journal of Microbiology and Infectious Diseases 1990;9:90–102.
Dentistry 2013;41:385–92. 48. Flannagan SE, Clewell DB, Sedgley CM. A retrocidal plasmid
31. Möller AJ. Microbiological examination of root canals and in Enterococcus faecalis: passage and protection. Plasmid
periapical tissues of human teeth. Methodological studies. 2008;59:217–30.
Odontologisk Tidskrift 1966;74:1–380. 49. Jungermann GB, Burns K, Nandakumar R, Tolba M, Venezia
32. Sundqvist G, Fidgor D, Sjogren U. Microbiology analysis of RA, Fouad AF. Antibiotic resistance in primary and
teeth with endodontic treatment and the outcome of persistent endodontic infections. Journal of Endodontics
conservative retreatment. Oral Surgery Oral Medicine Oral 2011;37:1337–44.
Pathology 1998;85:86–93. 50. Palmer KL, Kos VN, Gilmore MS. Horizontal gene transfer
33. Zoletti GO, Siqueira Jr JF, Santos KRN. Identification of and the genomics of enterococcal antibiotic resistance.
Enterococcus faecalis in root-filled teeth with or without Current Opinion in Microbiology 2010;13:632–9.
786 journal of dentistry 41 (2013) 779–786

51. Dunavant TR, Regan JD, Glickman GN, Solomon ES, formation by Enterococcus faecalis. Infection and Immunity
Honeyman AL. Comparative evaluation of endodontic 2004;72:6032–9.
irrigants against Enterococcus faecalis biofilms. Journal of 56. Hancock LE, Perego M. The Enterococcus faecalis fsr two-
Endodontics 2006;32:527–31. component system controls biofilm development through
52. Norrington DW, Ruby J, Beck P, Eleazer PD. Observations of production of gelatinase. Journal of Bacteriology
biofilm growth on human dentin and potential destruction 2004;186:5629–39.
after exposure to antibiotics. Oral Surgery Oral Medicine Oral 57. Shankar N, Baghdayan AS, Gilmore MS. Modulation of
Pathology Oral Radiology and Endodontics 2008;105:526–9. virulence within a pathogenicity island in vancomycin-
53. Wade WG. New aspects and new concepts of maintaining resistant Enterococcus faecalis. Nature 2002;417:746–50.
microbiological health. Journal of Dentistry 2010;38:21–5. 58. Di Rosa R, Creti R, Venditti M, D’Amelio R, Arciola CR,
54. Coburn PS, Gilmore MS. The Enterococcus faecalis cytolysin: a Montanaro L, et al. Relationship between biofilm
novel toxin active against eukaryotic and prokaryotic cells. formation, the enterococcal surface protein (Esp) and
Cellular Microbiology 2003;5:661–9. gelatinase in clinical isolates of Enterococcus faecalis and
55. Tendolkar PM, Baghdayan AS, Gilmore MS, Shankar N. Enterococcus faecium. FEMS Microbiology Letters
Enterococcal surface protein, Esp, enhances biofilm 2006;256:145–50.

You might also like