Skip to main content

Comparative evaluation of antimicrobial efficacy of different combinations of calcium hydroxide against Enterococcus faecalis

Abstract

Background

The study aims to compare the synergistic antibacterial efficacy of different combinations of calcium hydroxide as an intracanal medicament against E. faecalis.

Material and methods

The current study included four hundred extracted human permanent mandibular premolar teeth. After complete chemo-mechanical preparation, the middle third of the root was sectioned using a rotary diamond disc and a total of 400 samples were obtained. The specimens were inoculated with E. faecalis for 21 days. After that, specimens were divided into five groups (n = 80) based on materials used for the disinfection of samples: Group I, calcium hydroxide alone; Group II, calcium hydroxide + 2% chlorhexidine gel; Group III, calcium hydroxide + 2% chitosan gel; Group IV, calcium hydroxide + 0.02% silver nanoparticle gel; Group V, calcium hydroxide + Bioactive glass S53P4. Dentin shavings from the apical third were obtained from the inner third of dentin were obtained using gates glidden no.1 to the apical depth, followed by no.2, 3, 4 and 5 analyzed for E. faecalis using the culture method. One-way analysis of variance (ANOVA) was used for data analysis, followed by post-hoc Tukey's test for multiple comparisons of means to check the difference in bacterial inhibition between the groups.

Results

ANOVA results revealed a significant reduction of bacterial counts in all the groups compared (p < 0.001). Intergroup comparison showed maximum bacterial reduction (p < 0.001) with calcium hydroxide + bioactive glass S53P4 compared with other groups.

Conclusion

Synergistic effect of calcium hydroxide showed better bacterial reduction compared to calcium hydroxide alone. Among the combinations evaluated, calcium hydroxide with bioactive glass, found to be most effective compared to other groups.

Peer Review reports

Introduction

Optimal reduction of the microbial load from the root canal system is claimed to be the crucial step for the successful endodontic treatment [1, 2]. Studies have demonstrated 40–60% cultivable bacteria within the root canal system even after sodium hypochlorite irrigation [3,4,5]. Primarily, when the endodontic infections are considered, E. feacalis has shown to be the primitive bacterial cause in apical periodontitis cases. The claimed gram-positive microorganism is also shown to exhibit resistance and seen to be persistent in secondary root canal infections [2]. The typical nature of this microbe is its ability to form a mono-species biofilm in the root canal system, without any additional support from the other microbial species [6]. Even the E. feacalis has also shown higher resistance to the chemo-mechanical debridement and other adjunctive therapies rendered during the root canal treatment [7].

Various changes in the treatment procedures have been applied in Endodontics to overcome the above-mentioned issues. Although, the success of the endodontic treatment is still mainly dependent on the instrumentation and root canal disinfection [8]. It is essential to optimally disinfect the root canal system, to prevent the inherent tissue damage caused by bacteria and their byproducts released into the periapical areas [9]. The data from the current studies claim the inefficiency of conventional chemo-mechanical debridement in eliminating the microbial load from the root canal system [8,9,10].

Calcium hydroxide (CH) is the most popular intracanal medicament in Endodontics. Chemically it’s a strong base that is in contact with the aqueous liquids [11]. It exerts its antimicrobial action by dissociation reaction. Highly oxidant anions such as calcium and hydroxyl are released into the targeted site, which show extreme reactivity to various biomolecules [12]. The released hydroxyl ions, through direct or indirect contact, induce antibacterial effect through diffusion into the entire root canal system. Over the years, CH is considered a gold standard intracanal medicament [13, 14], but there is an ambiguity in its efficiency against Enterococcus species, mainly when colonized within the dentinal tubules [14]. E. faecalis is resistant to the alkaline pH which hinders the potentiality of the calcium hydroxide. E. faecalis tends to regulate its environmental pH by its proton pump property allowing its survival and perpetuating the infection. The previous report has revealed E. faecalis to be resistant and can adapt according to environmental change and penetrate the dentinal tubules. CH intracanal medicament is not equally effective for the bacterial types, inhabited in the root canal system. Moreover, the CH can be inactivated by dentin exudate evident from the periapical area [15,16,17].

Therefore, several studies focused on reporting the combination of medicaments to minimize the formation of resistant bacterial strain and eventually promote a synergistic effect with higher antibacterial efficacy [18,19,20,21,22]. Indeed, different agents, such as chlorhexidine [18], chitosan [19], silver nanoparticle [20], and bioactive glass [21], present a synergic antibacterial effect with CH. Chlorhexidine (CHX) has been claimed to be used as an potent intracanal medicament, both in vital and non-vital cases [22]. Furthermore, CHX intracanal medicament is claimed to be more effective than a CH when used as a dressing for eliminating E. feacalis [18]. Even though the mixture of CH and CHX is found to generate reactive oxygen species, the literature report stated no trace of parachloroaniline on the combination. This was mainly due to the immediate disintegration of CHX molecule which prevented the precipitate formation further [23].

Chitosan, a natural polysaccharide, is a copolymer of glucosamine and N-acetyl glucosamine. It has shown to be effective against a wide range of fungus and bacteria [24]. Previous studies were extensively conducted on chitosan as an intracanal medicament and tested its effectiveness towards various root canal pathogens [25, 26]. However, reports are lacking on the CH and chitosan combinations [19, 27], and testing its efficacy against the specific root canal pathogens.

Nanoparticles are nano-scale smaller sized molecules, with larger surface area and higher surface reactivity. Nanoparticles claimed to be alternative options in the modern dentistry, for both the prevention and the therapeutic benefit too [28, 29]. Silver nanoparticle (AGNP) has gained popularity in both restorative dentistry and endodontics as an additive in restorative materials [30, 31], as a dental adhesive [32], as an irrigant [33], intracanal medicament [34] and also as an additive antibacterial component of the root canal sealers [35]. Over the years, AGNP has been used as an intracanal medicament due to their strong antibacterial effect. They are claimed to be effective against gram-positive and negative bacteria [36] and multidrug-resistant bacteria [37]. Nevertheless, AGNP present some degree of toxicity depend on the concentration of the agent. Literature reported that these variations could be because of the differences in the size, dose, concentration, duration of action and the studied cell lines in various studies [38]. So, noteworthy is to study the critical AGNP concentration, alone or in combination with CH, which could induce a therapeutic benefit [20]. Hence, in the current study, we have planned to assess the real time benefit of combination of silver nanoparticles with CH as an intracanal medicament.

Bioactive glass (BAG) is a silica-based glass. The first-generation BAG is the 45S5. The composition of 45S5 is 45% SiO2, 24.5% CaO, 24.5% Na2O, 6% P2O5 [39]. The later developed compound with variation is the BAG S53P4. It is composed of SiO2 (53%), Na2O (23%), CaO (20%), P2O4 (4%) [40]. The BAG antibacterial property is dependent on the particle size. The smaller the size greater is the surface exposure of alkaline material to the liquid medium [41]. BAG has only been popular in recent days as an intracanal medicament. Its major advantage, when used as an intracanal medicament, is its antimicrobial ability along with an additional ion releasing capability, which helps in the alkalization of the medium [21]. There are several mechanisms explaining the mode of BAG action when used as an intracanal medicament. The accepted mechanism being the changes in the osmotic pressure, environmental pH, and needle like sharp debris produced by the materials, leads to the microbial cell damage, ultimately helping in the easier penetration of the antimicrobial components into the cytoplasm [42]. Although, growing evidence is in favor of using BAG as an intracanal medicament, with various studies focusing on its usage by altering the compositions and incorporating various chemical combinations.

Hence, the present study aimed to compare the synergistic antibacterial efficacy of different combinations of CH as an intracanal medicament against E. faecalis. The novelity of the current study lies in the different interventions namely CH, CHX, Chitosan, AGNP gel, and BAG 45S5 compared altogether. The null hypothesis states that there is no difference in the reduction of the E.faecalis count among the five different intracanal medicaments tested in the current study.

Materials and methods

The present in vitro study was conducted after obtaining the ethical approval from the institutional ethical committee (IHEC/SDC/FACULTY/21/ENDO/133).

Preparation of dentin specimen

Four hundred extracted mandibular premolars with mature apices were incorporated in the study. The inclusion criteria for the selected specimens were teeth with single root and single root canal, intact teeth with no caries, cracks or root resorptions. Teeth with root caries, curved canals and canal calcifications were excluded. The single canal morphology was confirmed with intraoral periapical radiographs taken at different angulations. The canal curvature was assessed using Schneider’s classification. Teeth with curvatures greater than 5° elicited through an intra-oral periapical radiograph, were excluded for the study. The initial apical diameter of the specimens was assessed by analyzing the CBCT sections. All the CBCT scanned specimens were assessed for their smallest diameter at 1 mm short of the root apex using an OnDemand 3D software (OnDemandedApp 1.0.9.2225; Cybermed, Inc, Seoul, South Korea) directly on axial sections, perpendicular to the canal. The entire assessment was done in an LCD monitor at (1366 × 768 pixels) resolution, to avoid selecting the specimens, whose diameter is more than the choosen preparation sizes in the current study. The current study protocol was similar to the previous literature [43,44,45].

An access preparation was performed with a high-speed round bur. After access cavity preparation, the patency of the canals was determined using K-files ISO # 10 (Dentsply Maillefer, Ballaigues, Switzerland). Only teeth whose canal width near the terminus was approx. comparable with ISO # 20 K-file were only included. After achieving the patency, the working length of the specimens was determined. After the working length confirmation, the instrumentation was carried out using protaper gold rotary files (Dentsply Maillefer, Ballaigues, Switzerland). The specimens were enlarged to a maximum size of 50/05 at the working length. In sue course of instrumentation, the irrigation was carried out using 10 ml of 3% sodium hypochlorite (Parcan, Septodont, India). A 30 guuage side vented needle irrigation (NaviTip, Ultradent Products, South Jordan, UT, USA) was carried out by placing the tip 1 mm short of the tentative working length. After the complete instrumentation, irrigation was carried out using 5 ml of 3% sodium hypochlorite and 3 ml of 17% ethylene diamine tertraacetic acid (MD Cleanser, Meta Biomed, India). The final rinse was carried out using saline and canals were dried using paper points.

After which, a rotary diamond disk was used to dissect the specimen to obtain 6 mm of the middle third of the root. The external diameter was standardized by removing the cementum from the root surface to 4 mm [46]. Then, using gates glidden drill no.3 (Mani Inc, Tochigi-ken, Japan), the internal diameter was standardized using a slow speed handpiece (NSK, Tokyo, Japan) till the apex. Finally, 3% sodium hypochlorite and 17% ethylene diamine tetraacetic acid were used for 5 min to remove the inoraganic and oragainc debris from the specimens. The final rinse was again carried out using saline and canals were dried using paper points. Next, the teeth were placed for 10 min in distilled water to remove the chemicals' remnant trace and then subjected to autoclave at 121 °C for 20 min [47].

Contamination of specimen

The E. faecalis (ATCC 29212) strain was used as a test organism in the current study. It was grown on tryptone soya agar (TSB), suspended in 5 mL of TS broth and incubated for 4 h at 37 °C. The turbidity was equivalent to 0.5 McFarland standard. A 50 μL of the E. faecalis inoculums were transferred into pre-sterilized microcentrifuge tubes containing 1 mL of tryptone soy broth. The dentin specimens were transferred into the fresh broth containing E. faecalis at the end of 24 h and maintained under laminar flow (Clean Air, Mumbai, India). The culture purity was confirmed by sub-culturing 5 μL of the broth from the incubated dentin specimens in TSB on tryptone soy agar plates. The samples were incubated with E. faecalis at 37 °C for 21 days [47].

Antimicrobial assessment

After 21 days of the incubation period, the specimens were rinsed with 5 mL of sterile water to remove the inoculated broth. Four hundred specimens were randomly allocated into 5 groups (n = 80) using a computer generated randomization procedure (www.random.org) and were assigned to the groups, based on the different intracanal medicament as follow:

  • Group I: CH (Ref no.23,923–2; SigmaAldrich, Mumbai, India);

  • Group II: CH + 2% CHX gel (Ref no.24800; Sigma-Aldrich);

  • Group III: CH + 2% chitosan gel (Everest Biotic, Bangalore, India);

  • Group IV: CH + 0.02% AGNP gel (huzheng nanotechnology co ltd);

  • Group V: CH + BAG S53P4 (AbminDent; Abmin Technologies Ltd, Turku, Finland).

The particle size for BAG S53P4 was 45 µm. For Group II, III, IV and V, sterile saline was used to mix the agents and adjust to a ratio of 1.5:1 to obtain paste-like consistency. The medicament in group 1 was mixed with sterile saline at a ratio of 1.5:1 [48]. The reason for adjusting the ratio was to standardaize the obtained mix as suggested in the previous similar study design [48]. Hence, in the current study, the agents were mixed and adjusted to a ratio of 1:5:1. The medicaments were then placed into the root canal, after which the ends of the specimen were sealed with temporary cement (cavit G;3 M ESPE, Seefeld, Germany) [47]. The samples were incubated in an anaerobic environment at 37 °C for seven days.

After seven days, the coronal seal was removed and the medicaments from canals were taken out using distilled water. Dentin shavings obtained from the inner third of dentin were obtained using gates glidden no.1 to the apical depth, followed by no. 2, 3, 4 and 5 [46, 47, 49, 50]. Before the experimentation, the initial weight of the uncapped Eppendorf tube was recorded in triplicate using an analytical balance (Sartorius Intec, Hamburg, Germany). The tubes were then finally incubated at 70 degree celcius for 5 days. The postinstrumentation dentin shavings were then weighed. Finally the dentinal shavings were immersed in 1 ml of TSA broth in 1.5 ml Eppendorf tubes. The tubes were vibrated in Fisher Vortex equipment for 2 min to homogenize the samples.

Culture methods for colony-forming units

A tenfold dilutions were prepared, and 1 ml of aliquots of suspensions were seeded on a Petri dish with 5% sheep blood TSA plates and then incubated at 37 °C for 48 h. The colony-forming units (CFU) grown were counted using a stereomicroscope, and log transformation was carried out for the values obtained.

Sample size and statistical analysis

A priori sample size calculation was done using G* power software (Universität Düsseldorf: Psychologie – HHU). Considering the ANOVA test (main effects and interactions) for comparing means of five study groups with an effect size (Cohen’s d value) of 0.25, confidence interval (1-β error) of 95% and 0.05 α, a total sample of 400 was calculated. With this sample size, the power of the study was estimated to be 0.95. Each study group has a sample size of 80. The data were statistically analyzed with a one-way analysis of variance (ANOVA), followed by post hoc Tukey's HSD multiple comparisons of means to check the difference in bacterial inhibition between the groups.

Results

The mean CFU for all the groups were Group I = 51.23 × 102, Group II = 35.23 × 102, Group III = 37.26 × 102, Group IV = 26.14 × 102, Group V = 23.25 × 102. ANOVA results revealed the statistical reduction (p < 0.001) in all the groups compared (Fig. 1). Intergroup comparison revealed that Group V was most effective (p < 0.001) against E. faecalis followed by Group IV, Group II and Group III. The least effective was Group I where CH was alone. The current study results have shown CH combinations to be more effective against E. feacalis as compared to the CH alone. Among the various combinations assessed, CH + BAG S53P4 combination has shown more reduction in the CFU counts after the usage of medicament. Whereas the CH + chitosan combintion was the least effective in reducing the CFU counts among all the combinations compared (Table 1).

Fig. 1
figure 1

Bar graph showing Intergroup comparative analysis of E. faecalis Counts

Table 1 Tukey’s Post-hoc analysis showing intergroup comparison of E. faecalis counts

Discussion

Endodontic treatment should focus on the elimination of bacterial biofilm from the root canal system [51]. Habitation of bacterial biofilm within the canal is the primary cause of pulpal and periapical disease [52]. Incomplete removal of these microorganisms during the endodontic treatment can result in infection, which tends to be majorly responsible for root canal treatment failures [53, 54]. Although, E. faecalis, is one of the most prevalent species found in canals of teeth with post treatment apical periodontitis, the current eveidence suggests that the post treamnet infections are usually mixed and caused by diufferent bacterial combinations [55].

Moreover, the root canal system's dentin composition and anatomical complexities in persistent infection tend to cause limitation to root canal disinfection [56]. In such a situation, chemical disinfection alone might not be sufficient for the complete elimination of microorganisms. Therefore, the choice of intracanal medicament plays a significant role in this condition. Haapasalo and Ørstavik proposed an in vitro model for the assessment of disinfection of dentinal tubules [57]. A modified model was used in the present study as we obtained the extracted teeth from humans compared to the previous ones, which used bovine teeth [46].

The present study intended to assess different agents in combination with CH when used as intraoperative root canal medicament. There are no reports on CH with BAG and CH with AGNP. The literature is not as specific on its usage, especially as an intracanal medicament or an inter-appointment dressing controlling the endodontic infections. As compared to the CH intracanal medicament, BAG is claimed to be superior in terms of its remineralisation ability and helpful in bone formation by hydoroxyapatite layer formation [21]. Hence, studies are more warranted on assessing the various benefits on using BAG as an intracanal medicament. As other medicaments, it also needs as an adequate vehicle for easier mixing and dispensing the medicament into the root canal system. The vehicle used for intracanal medicament, should also support for ion dissolution, solubility and reabsorption. Distilled water and saline are the ideal vehicles for BAG when used as an intracanal medicament [58]. The literature evidence from the ex-vivo and in vitro studies is supportive for the its immense biocompatibility, osseo-induction and osseo-conduction ability [59]. The results of the present study have revealed 23.25 × 102 bacterial reduction with CH + BAG S53P4 followed by 26.14 × 102 bacterial reduction with CH + 0.02% AGNP gel. However, CH + 2% CHX gel and CH + 2% chitosan gel had an overall reduction of only 35.23 × 102 and 37.26 × 102 respectively. Therefore, the hypothesis null was rejected since there was a statistical differences between the groups. In addition, the results revealed the most negligible bacterial reduction when CH was used alone, without any combinations.

BAG S53P4 has shown significant bacterial reduction. However, the mechanism of action is not fully understood. The possible activity might be due to the pH increase and osmotic pressure of the aqueous BAG that cause surface deterioration due to Na, Si, PO4 ions, which eventually causes the death of microorganisms [40, 60].

Studies have also shown the particle size of BAG can contribute to surface deterioration [41]. BAG S53P4 has a particle size of 45 µm. Therefore, this can also be a reason for good antibacterial activity.

The first study by Zehnder et al. assessed the antibacterial efficacy of BAG S53P4 compared with CH and reported CH as ineffective compared with a BAG [40]. On the other hand, Mehrvarzfar et al. reported CH to be superior to BAG 45S5 [61]. In contrast, Krithika datta et al. reported no significant difference between both the medicaments. Also, antibacterial efficacy was reported to increase with an increase in time [40, 48].

Krithika datta et al. showed 2% CHX gel to be more effective than BAG [48]. However, the present study results revealed that a combination of CH + BAG S53P4 is more effective in reducing the bacterial load than CH + 2% CHX gel. Studies reported to date have not assessed the combined efficacy of CH with BAG over other agents such as chitosan and AGNP.

Regarding CH and CH + 2% CHX gel, the authors of the present study have reported CH + 2% CHX gel to show a better antibacterial reduction. The reason is the interaction of the positive charge of the CHX with the phosphate groups on microbial cell walls, which is negatively charged, thereby altering the osmotic equilibrium of the cell. Eventually, cell wall permeability increases, enabling the CHX molecule to penetrate the bacteria [47]. Previous studies reported similar results, which showed CHX to be superior to CH [48, 62, 63].

Its substantivity, protein-binding and least cytotoxic properties have also been well documented in the literature [64]. Basically CHX is a bis-biguanide, which exhibits a bacteriostatic effect at lower concentration and bactericidal effect at higher concentrations. The optimal antibacterial effect of CHX is achieved at a pH of 5.5 to 7 [18]. The ideal properties of CHX as an intracanal medicament are mainly exhibited due to its alkaline pH [65]. It has various benefits when used as a medicament. It is claimed to neutralize the remnant organic tissue debris in the root canal after chemo-mechanical debridement [66]. It induces an osteo-promotive environment by continuous release of hydroxyl ions, leading to increased osteogenic activity [65]. Its additional benefits include the neutralization of bacterial cell walls, especially the lipopolysaccharide, and also help in better disinfection of the root canal system [67]. The toxic effects on the bacterial cell wall are mainly achieved by the interaction of the positively charged molecule of CHX with the negatively charged bacterial cell wall. There are reports claiming the increased antimicrobial activity of CHX, when combined with CH [68]. Usually, the combination aims at increasing the antimicrobial activity of CH while maintaining its biological and mechanical properties [68]. The literature claims the additive antimicrobial effect is not due to the CHX component alone. But the research claims the adjunctive effect is mainly due to the realse of various byproducts from the CHX, by fragmentation process. As these byproducts tend to have a higher pH, they exhibit both as an antioxidant and pro-oxidant, thereby enhancing its effects.

Savita et al. reported that both agents showed the highest bacterial reduction against E. faecalis in case of persistent infections [69]. Literature evidence has also shown that 2% CHX to be more efficient in bacterial reduction than 2% chitosan [70]. The reason might be due to the increased diffusion of CHX into the dentinal tubules [48, 71, 72]. Also, CHX was found to have increased penetration into dentinal tubules compared to chitosan [73].

Perochena et al. demonstrated the addition of chitosan nanoparticles to CH significantly had better antibacterial efficacy than CH when used alone [74]. The reason might be due to the enhanced stability of calcium ion binding to the surface of chitosan nanoparticles. Similar results were also observed in the study by Archana et al. [70].

It has also been reported as being biocompatible and biodegradable, and with suitable antibacterial properties. Furthermore, the chitosan is claimed to have a higher surface area and charge density and react with the bacterial cell surface, eventually destroying the bacterial cells [75]. Chitosan has an immense potential in inhibiting the biofilm formation. Its ant biofilm activity is mainly due to interaction occurring with microbial cellular and sub-cellular components [71]. The major advantage with chitosan is its higher biocompatibility and its lower cytotoxicity making it an ideal material to be used as an intracanal medicament.

Charannya et al. reported a combination of CHX and AGNP showed higher antibacterial activity than individual compounds [76]. The antimicrobial action against bacterial cells might vary depending on the characteristics of nanoparticles, such as contact time, concentration, particle size and surface charge [34].

Contact time for AGNP plays a vital role in eliminating microorganisms from the root canal system. The antibacterial activity is due to the interaction of AGNP with the bacterial cell wall, which causes structural changes leading to the destruction of tissue protein [37]. It also has an increased surface area to volume ratio, which increases the reactivity [77]. The antibacterial property was reported to be enhanced with the smaller particle size of approximately 1–100 nm, increasing the chemical reactivity [37]. AGNP have a broader spectrum antibacterial and anti-biofilm activity and due to their smaller size they are able to penetrate the complex areas of the root canal system.

It is said to be biocompatible when used at a lower concentration [78]. In vitro study data reported the increased oxidative stress, cytotoxicity and pulmonary inflammation with the larger sized AGNP as compared with the smaller ones [79]. There is always a controversial report on the evident cytotoxic nature of these nanoparticles. Indeed, studies have shown that AGNP not be suggested as root canal irrigant [29, 80].

Rodrigues et al. had reported no significant difference between AGNP and CHX. The present study result revealed that a combination of CH with CHX showed reduced antibacterial efficacy compared with CH with a AGNP [33].

The present study results correlated with Afkhami et al., wherein CH with AGNP showed better antibacterial efficacy than CH with CHX [81]. Also, CH with AGNP showed better bacterial reduction when compared to CH alone, which is also similar to our study results.

When the limitations of the present study have to be considered, CFU assessing the E. faecalis might not indeed translate a clinical condition. The main limitation of the study is that, it did not employ biofilm analysis rather than study assessed only the planktonic culture. The complex root canal anatomy houses the infected tissue along with the biofilm masses. Ideally, the root canal biofilm is extremely complex and organized. Its almost a difficult task or impossible to duplicate its characteristics in the invitro experimentations. Studies on mono or dual species over simplify the real root canal ecology and might not give a true implication of the clinically achievable results [82]. Hence, future studies are advised to be carried out by considering all these factors.

Conclusions

Combination with calcium hydroxide showed higher antibacterial efficacy than when used alone. Amongst which calcium hydroxide with bioactive glass showed better antimicrobial efficiency against E. faecalis when compared to other combinations of calcium hydroxide.

Availability of data and materials

The data will be available on reasonable request from the corresponding author.

References

  1. Alghamdi F, Shakir M. The influence of enterococcus faecalis as a dental root canal pathogen on endodontic treatment: a systematic review. Cureus. 2020. https://doi.org/10.7759/cureus.7257.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Barbosa-Ribeiro M. Microbiological investigation in teeth with persistent/secondary endodontic infection in different stages of endodontic retreatment. Eur Endod J. 2020. https://doi.org/10.14744/eej.2020.73626.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Amato M, Pantaleo G, Abtellatif D, Blasi A, Gagliani M, Iandolo A. An in vitro evaluation of the degree of pulp tissue dissolution through different root canal irrigation protocols. J Conserv Dent. 2018;21:175–9. https://doi.org/10.4103/JCD.JCD_49_18.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Minervini G, Franco R, Marrapodi MM, Ronsivalle V, Shapira I, Cicciù M. Prevalence of temporomandibular disorders in subjects affected by parkinson disease: a systematic review and metanalysis. J Oral Rehabil. 2023. https://doi.org/10.1111/joor.13496.

    Article  PubMed  Google Scholar 

  5. Teja KV, Janani K, Srivastava KC, Shrivastava D, Jose J, Marya A, Karobari MI. Comparison of herbal agents with sodium hypochlorite as root canal irrigant: a systematic review of in vitro studies. Evid Based Complement Alternat Med. 2021;2021:1–11. https://doi.org/10.1155/2021/8967219.

    Article  Google Scholar 

  6. Cosan G, Ozverel CS, Yigit Hanoglu D, Baser KHC, Tunca YM. Evaluation of antibacterial and antifungal effects of calcium hydroxide mixed with two different essential oils. Molecules. 2022;27:2635. https://doi.org/10.3390/molecules27092635.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Estrela C, Silva JA, Alencar AHGD, Leles CR, Decurcio DA. Efficacy of sodium hypochlorite and chlorhexidine against enterococcus faecalis: a systematic review. J Appl Oral Sci. 2008;16:364–8. https://doi.org/10.1590/S1678-77572008000600002.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Di Stasio D, Romano A, Gentile C, Maio C, Lucchese A, Serpico R, Paparella R, Minervini G, Candotto V, Laino L. Systemic and topical photodynamic therapy (PDT) on oral mucosa lesions: an overview. J Biol Regul Homeost Agents. 2018;32(2 Suppl. 1):123–6.

  9. Rathi S, Chaturvedi S, Abdullah S, Rajput G, Alqahtani NM, Chaturvedi M, Gurumurthy V, Saini R, Bavabeedu SS, Minervini G. Clinical Trial to Assess Physiology and Activity of Masticatory Muscles of Complete Denture Wearer Following Vitamin D Intervention. Medicina (Kaunas). 2023;59(2):410. https://doi.org/10.3390/medicina59020410.

  10. Rico-Romano C, Zubizarreta-Macho A, Baquero-Artigao M, Mena-Alvarez J. An analysis in vivo of intracanal bacterial load before and after chemo-mechanical preparation: a comparative analysis of two irrigants and two activation techniques. J. Clin. Exp. Dent. 2015; 0–0, https://doi.org/10.4317/jced.52585.

  11. Minervini G, Lucchese A, Perillo L, Serpico R, Minervini G. Unilateral superior condylar neck fracture with dislocation in a child treated with an acrylic splint in the upper arch for functional repositioning of the mandible. Cranio. 2017;35(5):337–41. https://doi.org/10.1080/08869634.2016.1203560.

  12. Abdellatif D, Amato A, Calapaj M, Pisano M, Iandolo A. A novel modified obturation technique using biosealers: An ex vivo study. J Conserv Dent. 2021;24(4):369–73. https://doi.org/10.4103/jcd.jcd_142_21.

    Article  PubMed  Google Scholar 

  13. Siqueirajr J, Rocas I, Souto R, Deuzeda M, Colombo A. Actinomyces species, streptococci, and enterococcus faecalis in primary root canal infections. J Endod. 2002;28:168–72. https://doi.org/10.1097/00004770-200203000-00006.

    Article  Google Scholar 

  14. Ballal V, Kundabala M, Acharya S, Ballal M. Antimicrobial action of calcium hydroxide, chlorhexidine and their combination on endodontic pathogens. Aust Dent J. 2007;52:118–21. https://doi.org/10.1111/j.1834-7819.2007.tb00475.x.

    Article  PubMed  Google Scholar 

  15. George S, Kishen A, Song P. The role of environmental changes on monospecies Biofilm formation on root canal wall by enterococcus faecalis. J Endod. 2005;31:867–72. https://doi.org/10.1097/01.don.0000164855.98346.fc.

    Article  PubMed  Google Scholar 

  16. Haapasalo HK, Siren EK, Waltimo TMT, Orstavik D, Haapasalo MPP. Inactivation of local root canal medicaments by dentine: an in vitro study. Int Endod J. 2000;33:126–31. https://doi.org/10.1046/j.1365-2591.2000.00291.x.

    Article  PubMed  Google Scholar 

  17. Wang Z, Shen Y, Ma J, Haapasalo M. The effect of detergents on the antibacterial activity of disinfecting solutions in dentin. J Endod. 2012;38:948–53. https://doi.org/10.1016/j.joen.2012.03.007.

    Article  PubMed  Google Scholar 

  18. Athanassiadis B, Abbott P, Walsh L. The use of calcium hydroxide, antibiotics and biocides as antimicrobial medicaments in endodontics. Aust Dent J. 2007;52:S64–82. https://doi.org/10.1111/j.1834-7819.2007.tb00527.x.

    Article  PubMed  Google Scholar 

  19. Nunes BD, Rosendo RA, Filho AA, Fook MV, de Sousa WJ, Barbosa RC, Pina HD, da Silva Neto JE, Amoah SK, et al. Chitosan-based biomaterial, calcium hydroxide and chlorhexidine for potential use as intracanal medication. Materials 2021; 14: 488, https://doi.org/10.3390/ma14030488.

  20. Algazlan AS, Almuraikhi N, Muthurangan M, Balto H, Alsalleeh F. Silver nanoparticles alone or in combination with calcium hydroxide modulate the viability, attachment, migration, and osteogenic differentiation of human mesenchymal stem cells. Int J Mol Sci. 2022;24:702. https://doi.org/10.3390/ijms24010702.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Grazziotin-Soares R, Dourado LG, Gonçalves BLL, Ardenghi DM, Ferreira MC, Bauer J, Carvalho CN. Dentin microhardness and sealer bond strength to root dentin are affected by using bioactive glasses as intracanal medication. Materials. 2020;13:721. https://doi.org/10.3390/ma13030721.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Souza-Filho FJD, Soares ADJ, Vianna ME, Zaia AA, Ferraz CCR, Gomes BP. Antimicrobial effect and ph of chlorhexidine gel and calcium hydroxide alone and associated with other materials. Braz. Dent. J. 2008; 19: 28–33, https://doi.org/10.1590/S0103-64402008000100005.

  23. Barbin LE, Saquy PC, Guedes DF, Sousa-Neto MD, Estrela C, Pécora JD. Determination of para-chloroaniline and reactive oxygen species in chlorhexidine and chlorhexidine associated with calcium hydroxide. J Endod. 2008;34:1508–14. https://doi.org/10.1016/j.joen.2008.08.032.

    Article  PubMed  Google Scholar 

  24. Del Carpio-Perochena A, Kishen A, Shrestha A, Bramante CM. Antibacterial properties associated with chitosan nanoparticle treatment on root dentin and 2 types of endodontic sealers. J Endod. 2015;41:1353–8. https://doi.org/10.1016/j.joen.2015.03.020.

    Article  PubMed  Google Scholar 

  25. Thienngern P, Panichuttra A, Ratisoontorn C, Aumnate C, Matangkasombut O. Efficacy of chitosan paste as intracanal medication against enterococcus faecalis and candida albicans biofilm compared with calcium hydroxide in an in vitro root canal infection model. BMC Oral Health. 2022;22:354. https://doi.org/10.1186/s12903-022-02385-x.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Raura N, Garg A, Arora A, Roma M. Nanoparticle technology and its implications in endodontics: a review. Biomater Res. 2020;24:21. https://doi.org/10.1186/s40824-020-00198-z.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Harshitha V, Ranjini M, Nadig R. Antibacterial efficacy of nisin, calcium hydroxide, and triple antibiotic paste in combination with chitosan as an intracanal medicament against enterococcus faecalis – an in vitro study. J Conserv Dent. 2022;25:504. https://doi.org/10.4103/jcd.jcd_125_22.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Song W, Ge S. Application of Antimicrobial Nanoparticles in Dentistry. Molecules. 2019;24:1033. https://doi.org/10.3390/molecules24061033.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Shrestha A, Kishen A. Antibacterial Nanoparticles in Endodontics: A Review. J Endod. 2016;42:1417–26. https://doi.org/10.1016/j.joen.2016.05.021.

    Article  PubMed  Google Scholar 

  30. Bapat RA, Chaubal TV, Joshi CP, Bapat PR, Choudhury H, Pandey M, Gorain B, Kesharwani P. An overview of application of silver nanoparticles for biomaterials in dentistry. Mater Sci Eng C. 2018;91:881–98. https://doi.org/10.1016/j.msec.2018.05.069.

    Article  Google Scholar 

  31. Espinosa-Cristóbal LF, Martínez-Castañón GA, Téllez-Déctor EJ, Niño-Martínez N, Zavala-Alonso NV, Loyola-Rodríguez JP. Adherence inhibition of streptococcus mutans on dental enamel surface using silver nanoparticles. Mater Sci Eng C. 2013;33:2197–202. https://doi.org/10.1016/j.msec.2013.01.039.

    Article  Google Scholar 

  32. Melo MAS, Cheng L, Zhang K, Weir MD, Rodrigues LKA, Xu HHK. Novel dental adhesives containing nanoparticles of silver and amorphous calcium phosphate. Dent Mater. 2013;29:199–210. https://doi.org/10.1016/j.dental.2012.10.005.

    Article  PubMed  Google Scholar 

  33. Rodrigues CT, De Andrade FB, De Vasconcelos LRSM, Midena RZ, Pereira TC, Kuga MC, Duarte MAH, Bernardineli N. Antibacterial properties of silver nanoparticles as a root canal irrigant against Enterococcus Faecalis biofilm and infected dentinal tubules. Int Endod J. 2018;51:901–11. https://doi.org/10.1111/iej.12904.

    Article  PubMed  Google Scholar 

  34. Wu D, Fan W, Kishen A, Gutmann JL, Fan B. Evaluation of the antibacterial efficacy of silver nanoparticles against enterococcus faecalis biofilm. J Endod. 2014;40:285–90. https://doi.org/10.1016/j.joen.2013.08.022.

    Article  PubMed  Google Scholar 

  35. Rücker VB, Balbinot GDS, Collares FM, De Araújo Neto VG, Giannini M, Leitune VCB. Synthesis of silver core‐shell nanoparticles and their influence on an experimental resin endodontic sealer: an in Vitro analysis. Int Endod J. 2023; 56:289–303. https://doi.org/10.1111/iej.13859.

  36. Guzman M, Dille J, Godet S. Synthesis and antibacterial activity of silver nanoparticles against Gram-Positive and Gram-negative bacteria. Nanomedicine Nanotechnol Biol Med. 2012;8:37–45. https://doi.org/10.1016/j.nano.2011.05.007.

    Article  Google Scholar 

  37. Rai MK, Deshmukh SD, Ingle AP, Gade AK. Silver nanoparticles: the powerful nanoweapon against multidrug-resistant bacteria: activity of silver nanoparticles against MDR bacteria. J Appl Microbiol. 2012;112:841–52. https://doi.org/10.1111/j.1365-2672.2012.05253.x.

    Article  PubMed  Google Scholar 

  38. Akter M, Sikder MdT, Rahman MdM, Ullah AKMA, Hossain KFB, Banik S, Hosokawa T, Saito T, Kurasaki M. A systematic review on silver nanoparticles-induced cytotoxicity: physicochemical properties and perspectives. J Adv Res. 2018;9:1–16. https://doi.org/10.1016/j.jare.2017.10.008.

    Article  PubMed  Google Scholar 

  39. Munukka E, Leppäranta O, Korkeamäki M, Vaahtio M, Peltola T, Zhang D, Hupa L, Ylänen H, Salonen JI, Viljanen MK, et al. Bactericidal effects of bioactive glasses on clinically important aerobic bacteria. J Mater Sci Mater Med. 2008;19:27–32. https://doi.org/10.1007/s10856-007-3143-1.

    Article  PubMed  Google Scholar 

  40. Zehnder M, Soderling E, Salonen J, Waltimo T. Preliminary evaluation of bioactive glass S53P4 as an endodontic medication in vitro. J Endod. 2004;30:220–4. https://doi.org/10.1097/00004770-200404000-00009.

    Article  PubMed  Google Scholar 

  41. Waltimo T, Brunner TJ, Vollenweider M, Stark WJ, Zehnder M. Antimicrobial effect of nanometric bioactive glass 45S5. J Dent Res. 2007;86:754–7. https://doi.org/10.1177/154405910708600813.

    Article  PubMed  Google Scholar 

  42. Drago L, Toscano M, Bottagisio M. Recent evidence on bioactive glass antimicrobial and antibiofilm activity: a mini-review. Materials. 2018;11:326. https://doi.org/10.3390/ma11020326.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Sujith IL, Teja KV, Ramesh S. Assessment of irrigant flow and apical pressure in simulated canals of single-rooted teeth with different root canal tapers and apical preparation sizes: an ex vivo study. J Conserv Dent. 2021;24:314–22. https://doi.org/10.4103/jcd.jcd_651_20.

    Article  PubMed  Google Scholar 

  44. Choudhari S, Teja KV, Ramesh S, Jose J, Janani K, Kumar R. Assessment of apical pressures at different automated irrigant flow rates: an ex vivo study based on computational fluid dynamic analysis. Braz Dent Sci. 2022; 25. https://doi.org/10.4322/bds.2022.e3463.

  45. Campello AF, Marceliano-Alves MF, Siqueira JF Jr, Marques FV, Guedes FR, Lopes RT, AlvarengaFO, Alves FRF. Determination of the initial apical canal diameter by the first file to bind or cone-beam computed tomographic measurements using micro-computed tomography as the gold standard: an ex vivo study in human cadavers. J Endod. 2019; 45: 619–622. https://doi.org/10.1016/j.joen.2019.01.020.

  46. Basrani B, Tjäderhane L, Santos JM, Pascon E, Grad H, Lawrence HP, Friedman S. Efficacy of chlorhexidine- and calcium hydroxide–containing medicaments against enterococcus faecalis in vitro. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endodontology 2003; 96: 618–624. https://doi.org/10.1016/S1079-2104(03)00166-5.

  47. Kandaswamy D, Venkateshbabu N, Gogulnath D, Kindo AJ. Dentinal tubule disinfection with 2% chlorhexidine gel, propolis, morinda citrifolia juice, 2% povidone iodine, and calcium hydroxide. Int Endod J. 2010;43:419–23. https://doi.org/10.1111/j.1365-2591.2010.01696.x.

    Article  PubMed  Google Scholar 

  48. Krithikadatta J, Indira R, Dorothykalyani AL. Disinfection of dentinal tubules with 2%, chlorhexidine, 2% metronidazole, bioactive glass when compared with calcium hydroxide as intracanal medicaments. J Endod. 2007;33:1473–6. https://doi.org/10.1016/j.joen.2007.08.016.

    Article  PubMed  Google Scholar 

  49. Choudhari S, Pradeep S, Teja KV. Antimicrobial efficacy of a novel automated irrigation device as compared to conventional needle irrigation against enterococcus faecalis: an in vitro study. Cureus. 2023; 15: 45200. https://doi.org/10.7759/cureus.45200.

  50. Teja KV, Janani K, Kaligotla VA, Harini K. Comparative antimicrobial efficacy of oregano oil, chlorhexidine, and sodium hypochlorite against Enterococcus faecalis: An: in vitro: study. Endodontol. 2021;33:97–101. https://doi.org/10.4103/endo.endo_124_20.

    Article  Google Scholar 

  51. Janani K, Ajitha P, Sandhya R, Teja K. Chemical constituent, minimal inhibitory concentration, and antimicrobial efficiency of essential oil from oreganum vulgare against enterococcus faecalis: an in vitro study. J Conserv Dent. 2019;22:538. https://doi.org/10.4103/JCD.JCD_80_19.

    Article  PubMed  Google Scholar 

  52. Kishen A. Advanced therapeutic options for endodontic biofilms: advanced therapeutic options for endodontic biofilms. Endod Top. 2010;22:99–123. https://doi.org/10.1111/j.1601-1546.2012.00284.x.

    Article  Google Scholar 

  53. Jose J, Teja KV, Janani K, Alam MK, Khattak O, Salloum MG, Magar SS, Magar SP, Rajeshkumar S, Palanivelu A, et al. Preparation of a novel nanocomposite and its antibacterial effectiveness against enterococcus faecalis—an in vitro evaluation. Polymers. 2022;14:1499. https://doi.org/10.3390/polym14081499.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Siqueirajr J, Rocas I. Exploiting molecular methods to explore endodontic infections: part 2—redefining the endodontic microbiota. J Endod. 2005;31:488–98. https://doi.org/10.1097/01.don.0000157990.86638.49.

    Article  Google Scholar 

  55. Siqueira JF Jr, Rôças IN. Present status and future directions: microbiology of endodontic infections. Int Endod J. 2022;55:512–30. https://doi.org/10.1111/iej.13677.

    Article  PubMed  Google Scholar 

  56. Gummuluri S, Kavalipurapu VT, Kaligotla AV. Antimicrobial efficacy of novel ethanolic extract of morinda citrifolia against enterococcus feacalis by agar well diffusion method and minimal inhibitory concentration- an invitro study. Braz Dent Sci. 2019;22:365–70.

    Article  Google Scholar 

  57. Haapasalo M, Ørstavik D. In Vitro Infection and of Dentinal Tubules. J Dent Res. 1987;66:1375–9. https://doi.org/10.1177/00220345870660081801.

    Article  PubMed  Google Scholar 

  58. Shetty S. An in-vitro evaluation of the ph change through root dentin using different calcium hydroxide preparations as an intracanal medicament. J Clin Diagn Res. 2014. https://doi.org/10.7860/JCDR/2014/9374.4950.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Fernandes HR, Gaddam A, Rebelo A, Brazete D, Stan GE, Ferreira JMF. Bioactive glasses and glass-ceramics for healthcare applications in bone regeneration and tissue engineering. Materials. 2018;11:2530. https://doi.org/10.3390/ma11122530.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Stoor P, Söderling E, Salonen JI. Antibacterial Effects of a Bioactive Glass Paste on Oral Microorganisms. Acta Odontol Scand. 1998;56:161–5. https://doi.org/10.1080/000163598422901.

    Article  PubMed  Google Scholar 

  61. Mehrvarzfar P, Akhavan H, Rastgarian H, Mohammadzade Akhlagi N, Soleymanpour R, Ahmadi A. An in vitro comparative study on the antimicrobial effects of bioglass 45S5 vs. calcium hydroxide on enterococcus faecalis. Iran Endod J. 2011; 6:29–33.

  62. Gomes BPFA, Souza SFC, Ferraz CCR, Teixeira FB, Zaia AA, Valdrighi L, Souza-Filho FJ. Effectiveness of 2% chlorhexidine gel and calcium Hydroxide against Enterococcus Faecalis in bovine root dentine in Vitro: antibacterial activity of chlorhexidine gel and calcium hydroxide. Int Endod J. 2003;36:267–75. https://doi.org/10.1046/j.1365-2591.2003.00634.x.

    Article  PubMed  Google Scholar 

  63. Siqueira JF, De Uzeda M. Intracanal medicaments: evaluation of the antibacterial effects of chlorhexidine, metronidazole, and calcium hydroxide associated with three vehicles. J Endod. 1997;23:167–9. https://doi.org/10.1016/S0099-2399(97)80268-3.

    Article  PubMed  Google Scholar 

  64. Tandan M, Hegde M, Hegde P. Effect of four different intracanal medicaments on the apical seal of the root canal system: a dye extraction study. Indian J Dent Res. 2014;25:607. https://doi.org/10.4103/0970-9290.147104.

    Article  PubMed  Google Scholar 

  65. Tronstad L, Andreasen JO, Hasselgren G, Kristerson L, Riis I. PH Changes in dental tissues after root canal filling with calcium hydroxide. J Endod. 1981;7:17–21. https://doi.org/10.1016/S0099-2399(81)80262-2.

    Article  PubMed  Google Scholar 

  66. Torneck CD, Moe H, Howley TP. The Effect of Calcium hydroxide on porcine pulp fibroblasts in vitro. J Endod. 1983;9:131–6. https://doi.org/10.1016/S0099-2399(83)80031-4.

    Article  PubMed  Google Scholar 

  67. Safavi KE, Nichols FC. Alteration of biological properties of bacterial lipopolysaccharide by calcium hydroxide treatment. J Endod. 1994;20:127–9. https://doi.org/10.1016/S0099-2399(06)80057-9.

    Article  PubMed  Google Scholar 

  68. Putri FT, Caesarendra W, Królczyk G, Glowacz A, Prawibowo H, Ismail R, Indrawati RT. Human Walking Gait Classification Utilizing an Artificial Neural Network for the Ergonomics Study of Lower Limb Prosthetics. Prosthesis. 2023;5(3):647–65. https://doi.org/10.3390/prosthesis5030046.

  69. Savitha A, SriRekha A, Vijay R, Champa C, Jaykumar T. An in vivo comparative evaluation of antimicrobial efficacy of chitosan, chlorhexidine gluconate gel and their combination as an intracanal medicament against enterococcus faecalis in failed endodontic cases using real time polymerase chain reaction (QPCR). Saudi Dent J. 2019; 31: 360–366. https://doi.org/10.1016/j.sdentj.2019.03.003.

  70. Ghadirinejad K, Day CW, Milimonfared R, Taylor M, Solomon LB, Hashemi R. Fretting Wear and Corrosion-Related Risk Factors in Total Hip Replacement: A Literature Review on Implant Retrieval Studies and National Joint Replacement Registry Reports. Prosthesis. 2023;5(3):774–91. https://doi.org/10.3390/prosthesis5030055.

  71. Arnould A, Hendricusdottir R, Bergmann J. The complexity of medical device regulations has increased, as assessed through data-driven techniques. Prosthesis. 2021;3:314–30. https://doi.org/10.3390/prosthesis3040029.

    Article  Google Scholar 

  72. Chhikara K, Gupta S, Bose D, Kataria C, Chanda A. Development and Trial of a Multipurpose Customized Orthosis for Activities of Daily Living in Patients with Spinal Cord Injury. Prosthesis. 2023;5(2):467–79. https://doi.org/10.3390/prosthesis5020032.

  73. Arathi G, Rajakumaran A, Divya S, Malathi N, Saranya V, Kandaswamy D. Comparison of penetrating depth of chlorhexidine and chitosan into dentinal tubules with and without the effect of ultrasonic irrigation. J Oral Maxillofac Pathol. 2019;23:389. https://doi.org/10.4103/jomfp.JOMFP_194_19.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Shaikh S, Jamdade B, Chanda A. Effects of Customized 3D-Printed Insoles in Patients with Foot-Related Musculoskeletal Ailments—A Survey-Based Study. Prosthesis. 2023;5(2):550–61. https://doi.org/10.3390/prosthesis5020038.

  75. Shi Z, Neoh KG, Kang ET, Wang W. antibacterial and mechanical properties of bone cement impregnated with chitosan nanoparticles. Biomaterials. 2006;27:2440–9. https://doi.org/10.1016/j.biomaterials.2005.11.036.

    Article  PubMed  Google Scholar 

  76. Inchingolo F, Tatullo M, Marrelli M, Inchingolo AM, Picciariello V, Inchingolo AD, Dipalma G, Vermesan D, Cagiano R. Clinical trial with bromelain in third molar exodontia. Eur Rev Med Pharmacol Sci. 2010;14(9):771–4.

  77. Adina S, Dipalma G, Bordea IR, Lucaciu O, Feurdean C, Inchingolo AD, Septimiu R, Malcangi G, Cantore S, Martin D, et al. Orthopedic joint stability influences growth and maxillary development: clinical aspects. J Biol Regul Homeost Agents. 2020;34:747–56. https://doi.org/10.23812/20-204-E-52.

    Article  PubMed  Google Scholar 

  78. Colombo S, Gallus S, Beretta M, Lugo A, Scaglioni S, Colombo P, Paglia M, Gatto R, Marzo G, Caruso S, Paglia L. Prevalence and determinants of early childhood caries in Italy. Eur J Paediatr Dent. 2019;20(4):267–73. https://doi.org/10.23804/ejpd.2019.20.04.02.

  79. Wang X, Ji Z, Chang CH, Zhang H, Wang M, Liao Y-P, Lin S, Meng H, Li R, Sun B, et al. Use of coated silver nanoparticles to understand the relationship of particle dissolution and bioavailability to cell and lung toxicological potential. Small. 2014;10:385–98. https://doi.org/10.1002/smll.201301597.

    Article  PubMed  Google Scholar 

  80. Samiei M, Farjami A, Dizaj SM, Lotfipour F. Nanoparticles for antimicrobial purposes in endodontics: a systematic review of in vitro studies. Mater Sci Eng C. 2016;58:1269–78. https://doi.org/10.1016/j.msec.2015.08.070.

    Article  Google Scholar 

  81. Afkhami F, Pourhashemi SJ, Sadegh M, Salehi Y, Fard MJK. Antibiofilm efficacy of silver nanoparticles as a vehicle for calcium hydroxide medicament against enterococcus faecalis. J Dent. 2015;43:1573–9. https://doi.org/10.1016/j.jdent.2015.08.012.

    Article  PubMed  Google Scholar 

  82. Neelakantan P, Romero M, Vera J, Daood U, Khan AU, Yan A, Cheung GSP. Biofilms in endodontics-current status and future directions. Int J Mol Sci. 2017;18:1748. https://doi.org/10.3390/ijms18081748.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

None

Funding

This research received no external funding.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization K.V.T.; K.J.; K.C.S.; D.S.; M.D.B.; M.C. and G.M.; methodology, K.V.T.; K.J.; V.N.; and D.S.; software, K.C.S.; and V.N.; formal analysis, K.C.S.;and K.C.S.; investigation, K.V.T.; and K.J.; data curation, K.C.S.; and D.S.; writing—original draft preparation, K.V.T.; K.C.S.; M.D.B.; M.C.; G.M. and D.S.; writing—review and editing, K.V.T.; K.J.; K.C.S.; D.S.; M.D.B.; M.C.; G.M. and V.N.; supervision, K.C.S.; funding acquisition, K.C.S. and V.N. administration: K.C.S. All authors have read and agreed to the published version of the manuscript. 

Corresponding authors

Correspondence to Kumar Chandan Srivastava, Deepti Shrivastava or Marco Di Blasio.

Ethics declarations

Ethics approval and consent to participate

The Saveetha Dental College Ethics Committee gave its approval to this endeavor (Registry Number IHEC/SDC/FACULTY/21/ENDO/133). The study was carried out in accordance with the Helsinki Declaration of 1975, as revised in 2000. Informed consent was obtained from all the participants and/or legal guardians for the study.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Teja, K.V., Janani, K., Srivastava, K.C. et al. Comparative evaluation of antimicrobial efficacy of different combinations of calcium hydroxide against Enterococcus faecalis. BMC Oral Health 23, 849 (2023). https://doi.org/10.1186/s12903-023-03552-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12903-023-03552-4

Keywords