scholarly journals Antimicrobial Activity of Protamine-Loaded Calcium Phosphates against Oral Bacteria

Materials ◽  
2019 ◽  
Vol 12 (17) ◽  
pp. 2816 ◽  
Author(s):  
Masashi Fujiki ◽  
Kodai Abe ◽  
Tohru Hayakawa ◽  
Takatsugu Yamamoto ◽  
Mana Torii ◽  
...  

Protamine is an antimicrobial peptide extracted from fish. In this study, we loaded protamine onto dicalcium phosphate anhydride (DCPA), a dental material. Protamine was loaded by stirring DCPA into a protamine solution. To explore the antimicrobial activity of the materials, we cultivated Streptococcus mutans on fabricated discs for 24 h. When S. mutans was cultivated on the discs under no sucrose conditions, the loaded protamine was not released, and the ratio of dead bacteria increased on the surface of P (125) DCPA (half of the saturated level of protamine (125 ppm protamine) was loaded). Aside from P (500) DCPA (saturated level of protamine was loaded), some protamine was released, and the number of planktonic bacteria in the supernatant decreased. Using medium containing 1% sucrose, the release of protamine was promoted from P (125) DCPA due to lowered pH. However, lowering of the pH decreased the antimicrobial activity of protamine. On the other hand, P (500) DCPA released protamine before the pH was lowered, and biofilm formation was inhibited. The loaded protamine expressed antimicrobial activity, both on the surface of the materials and in the surrounding environment. The interaction of loaded protamine with calcium phosphates could promote the application of protamine in the dental field.

2020 ◽  
Vol 4 (s1) ◽  
pp. 121-121
Author(s):  
Zhou Ye ◽  
Joseph R. Rahimi ◽  
Nicholas G. Fischer ◽  
Hooi Pin Chew ◽  
Conrado Aparicio

OBJECTIVES/GOALS: Peri-implantitis is the inflammation of peri-implant mucosa and subsequent loss of supporting bone. Its treatment is only <40% successful mainly due to persistent bacterial infection. The goal of this project is to increase success rates by developing a robust antibiofilm multi-biomolecular membrane that can be placed around implant surfaces. METHODS/STUDY POPULATION: A collagen membrane was soaked in the antimicrobial peptide GL13K solution overnight to form an interpenetrating fibrillary network. The nanostructure of the membrane was imaged with scanning electron microscope (SEM). The hydrophobicity of the membrane was analyzed by water contact angle (WCA) measurements. The biodegradability was tested in a 0.01 mg/mL Type I collagenase solution for up to 5 weeks. The antimicrobial activity of the membrane was assessed with Gram-positive oral bacteria Streptococcus gordonii. The cytotoxicity was evaluated by culturing human gingival fibroblasts (HGF), and the osteogenesis was assessed using preosteoblasts MC3T3. Pure collagen membrane was used as the control. Statistical significance (p<0.05) was determined by one-way ANOVA with Tukey’s HSD test. RESULTS/ANTICIPATED RESULTS: The antimicrobial peptide GL13K self-assembled to short fibrils (< 1 µm long), which entangled with the larger collagen fibers (around 200 nm in diameter). The collagen fibers presented characteristic periodic banding structures, which provided biomimetic cues for cell behavior as extracellular matrix. The interpenetrated GL13K fibrils turned the highly hydrophilic collagen membrane to a hydrophobic membrane (WCA = 135 °) and significantly reduced the rate of degradation by collagenases. The developed membrane was efficient in preventing the attachment of S. gordonii. A large portion of the attached bacteria was killed on the surface of the membrane. The incorporation of GL13K did not affect the cytocompatibility of the membrane for HGF. DISCUSSION/SIGNIFICANCE OF IMPACT: We developed an antibiofilm membrane with interpenetrating collagen and antimicrobial peptide fibrils. The strong antimicrobial activity and low cytotoxicity support its further translational evaluation as scaffolds for increasing success rate in treating peri-implantitis.


Pharmaceutics ◽  
2020 ◽  
Vol 12 (4) ◽  
pp. 352
Author(s):  
Jan-Luca Schmid ◽  
Martin Kirchberg ◽  
Sandra Sarembe ◽  
Andreas Kiesow ◽  
Anton Sculean ◽  
...  

Periodontal therapy using antimicrobials that are topically applied requires slow or controlled release devices. The in vitro antimicrobial activity of biodegradable polymer formulations that contain a new minocycline lipid complex (P-MLC) was evaluated. The new P-MLC formulations that contained 11.5% minocycline were compared with pure minocycline or an existing commercial formulation, which included determination of minimal inhibitory concentration (MIC) values against two oral bacteria and activity on six-species periodontal biofilm. Moreover, the flow of gingival crevicular fluid (GCF) was modeled up to 42 days and the obtained eluates were tested both for MIC values and inhibiting biofilm formation. In general, MICs of the P-MLC formulations were slightly increased as compared with pure minocycline. Biofilm formation was clearly inhibited by all tested formulations containing minocycline with no clear difference between them. In 3.5 day old biofilms, all formulations with 250 µg/mL minocycline decreased bacterial counts by 3 log10 and metabolic activity with no difference to pure antimicrobials. Eluates of experimental formulations showed superiority in antimicrobial activity. Eluates of one experimental formulation (P503-MLC) still inhibited biofilm formation at 28 days, with a reduction by 1.87 log10 colony forming units (CFU) vs. the untreated control. The new experimental formulations can easily be instilled in periodontal pockets and represent alternatives in local antimicrobials, and thus warrant further testing.


2020 ◽  
Vol 202 (12) ◽  
Author(s):  
Delphine Dufour ◽  
Abdelahhad Barbour ◽  
Yuki Chan ◽  
Marcus Cheng ◽  
Taimoor Rahman ◽  
...  

ABSTRACT Bacteriocins are ribosomally synthesized proteinaceous antibacterial peptides. They selectively interfere with the growth of other bacteria. The production and secretion of bacteriocins confer a distinct ecological advantage to the producer in competing against other bacteria that are present in the same ecological niche. Streptococcus mutans, a significant contributor to the development of dental caries, is one of the most prolific producers of bacteriocins, known as mutacins in S. mutans. In this study, we characterized the locus encoding mutacin B-Ny266, a lantibiotic with a broad spectrum of activity. The chromosomal locus is composed of six predicted operon structures encoding proteins involved in regulation, antimicrobial activity, biosynthesis, modification, transport, and immunity. Mutacin B-Ny266 was purified from semisolid cultures, and two inhibitory peptides, LanA and LanA′, were detected. Both peptides were highly modified. Such modifications include dehydration of serine and threonine and the formation of a C-terminal aminovinyl-cysteine (AviCys) ring. While LanA peptide alone is absolutely required for antimicrobial activity, the presence of LanA′ enhanced the activity of LanA, suggesting that B-Ny266 may function as a two-peptide lantibiotic. The activation of lanAA′ expression is most likely controlled by the conserved two-component system NsrRS, which is activated by LanA peptide but not by LanA′. The chromosomal locus encoding mutacin B-Ny266 was not universally conserved in all sequenced S. mutans genomes. Intriguingly, the genes encoding LanAA′ peptides were restricted to the most invasive serotypes of S. mutans. IMPORTANCE Although dental caries is largely preventable, it remains the most common and costly infectious disease worldwide. Caries is initiated by the presence of dental plaque biofilm that contains Streptococcus mutans, a species extensively characterized by its role in caries development and formation. S. mutans deploys an arsenal of strategies to establish itself within the oral cavity. One of them is the production of bacteriocins that confer a competitive advantage by targeting and killing closely related competitors. In this work, we found that mutacin B-Ny266 is a potent lantibiotic that is effective at killing a wide array of oral streptococci, including nearly all S. mutans strains tested. Lantibiotics produced by oral bacteria could represent a promising strategy to target caries pathogens embedded in dental plaque biofilm.


2020 ◽  
Author(s):  
Ghada H Naguib ◽  
Mohamed T Hamed ◽  
Abdulkarim A Hatrom ◽  
Abeer M Alnowaiser ◽  
Sahar MN Bukhary ◽  
...  

Abstract Background The addition of antibacterial agents to oral prophylaxis materials showed inhibition of plaque accumulation and bacterial acid production. This study aimed to test the antimicrobial effect of distinctive oral mouth washes on an exemplary of oral bacteria after the addition of zein-coated MgO nanoparticles.Materials and Methods Three hundred and twelve groups were used in this study. MgO nanoparticles in five concentrations with zein and without zein-coating, were incorporated in three oral mouth washes: Listerine Zero, Listerine Total Control and Oral B in the mass percentages of 0.3%, 0.5%, 1%, 2%, 5% and 10%, in addition to controls with no MgO nanoparticles. The three mouthwashes with variable concentrations were studied in opposition of Staphylococcus aureus, Streptococcus mutans, Enterococcus faecalis and Candida albicans using two different tests, the Disc Diffusion Test (DDT) and Direct Contact Test (DCT). Data was analyzed with Kruskall-Wallis and Mann-Whitney U tests.Results Results showed a highly significant statistical difference of antimicrobial activity for all tested mouth washes with Zein coated MgO nanoparticles on Streptococcus mutans, Staphylococcus aureus, Enterococcus faecalis, and Candida albicans in the disc diffusion test. While in the DCT, all tested mouth washes with MgO nanoparticles with and without zein coating showed antimicrobial activity on all tested microorganisms.Conclusion Zein coated MgO nanoparticles is a potent antimicrobial agent when added to oral mouth washes. It is recommended to incorporate Zein MgO nanoparticles to Oral mouth washes to improve its antibacterial property.


2016 ◽  
Vol 50 (1) ◽  
pp. 48-61 ◽  
Author(s):  
Huanxin Tu ◽  
Yingying Fan ◽  
Xueping Lv ◽  
Sili Han ◽  
Xuedong Zhou ◽  
...  

Controlling the growth of cariogenic microorganisms such as oral streptococci is an adjunct therapy for caries-active individuals to prevent and treat caries. Here we investigated the antimicrobial activity of the synthetic amphipathic α- helical antimicrobial peptide GH12 (GLLWHLLHHLLH-NH2) against oral streptococci in vitro. Circular dichroism studies showed that GH12 takes on an α-helical conformation in the presence of membrane-mimicking solvents, and reversed-phase high-performance liquid chromatography studies showed that GH12 remains stable in saliva. The peptide showed bactericidal activity against oral streptococci, with minimum inhibitory concentrations ranging from 6.7 to 32.0 μg/ml. GH12 concentrations 4-fold higher than the minimum bactericidal concentration completely killed oral streptococci within 20 min. Treating oral streptococci with GH12 caused noticeable changes in bacterial viability and morphology based on confocal laser scanning microscopy and scanning electron microscopy. Effects of GH12 on biofilm formation and on viability of mature biofilm were quantified by crystal violet staining and the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. GH12 effectively inhibited biofilm formation and metabolic activity in biofilms of oral streptococci, especially S. mutans, S. sobrinus and S. salivarius. These results suggest that GH12 shows rapid and strong antimicrobial activity against oral streptococci in vitro, opening the door to preclinical and clinical studies to explore its potential for caries prevention and treatment.


2018 ◽  
Vol 84 (24) ◽  
Author(s):  
Wentao Jiang ◽  
Yufei Wang ◽  
Junyuan Luo ◽  
Xinwei Li ◽  
Xuedong Zhou ◽  
...  

ABSTRACTDental caries is a biofilm-mediated disease that occurs when acidogenic/aciduric bacteria obtain an ecological advantage over commensal species. In previous studies, the effects of the antimicrobial peptide GH12 on planktonic bacteria and monospecies biofilms were confirmed. The objectives of this study were to investigate the effects of GH12 on a cariogenic multispecies biofilm and to preliminarily explain the mechanism. In this biofilm model,Streptococcus mutansATCC 70061 was the representative of cariogenic bacteria, whileStreptococcus gordoniiATCC 35105 andStreptococcus sanguinisJCM 5708 were selected as healthy microbiota. The results showed that GH12 was more effective in suppressingS. mutansthan the other two species, with lower MIC and minimal bactericidal concentration (MBC) values among diverse type strains and clinical isolated strains. Therefore, GH12, at no more than 8 mg/liter, was used to selectively suppressS. mutansin the multispecies biofilm. GH12 at 4 mg/liter and 8 mg/liter reduced the cariogenic properties of the multispecies biofilm in biofilm formation, glucan synthesis, and lactic acid production. In addition, GH12 suppressedS. mutanswithin the multispecies biofilm and changed the bacterial composition. Furthermore, 8 mg/liter GH12 showed a selective bactericidal impact onS. mutans, and GH12 promoted hydrogen peroxide production inS. sanguinisandS. gordonii, which improved their ecological advantages. In conclusion, GH12 inhibited the cariogenic properties and changed the composition of the multispecies biofilm through a two-part mechanism by which GH12 directly suppressed the growth ofS. mutansas well as enhanced the ecological competitiveness ofS. sanguinisandS. gordonii.IMPORTANCEDental caries is one of the most prevalent chronic infectious diseases worldwide, with substantial economic and quality-of-life impacts.Streptococcus mutanshas been considered the principal pathogen of dental caries. To combat dental caries, an antimicrobial peptide, GH12, was designed, and its antibacterial effects on planktonicS. mutansand the monospecies biofilm were confirmed. As etiological concepts of dental caries evolved to include microecosystems, the homeostasis between pathogenic and commensal bacteria and a selective action on cariogenic virulence have increasingly become the focus. The novelty of this research was to study the effects of the antimicrobial peptides on a controlled cariogenic multispecies biofilm model. Notably, the role of an antimicrobial agent in regulating interspecific competition and composition shifts within this multispecies biofilm was investigated. With promising antibacterial and antibiofilm properties, the use of GH12 might be of importance in preventing and controlling caries and other dental infections.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Zheng Wang ◽  
Yujie Zhou ◽  
Qi Han ◽  
Xingchen Ye ◽  
Yanyan Chen ◽  
...  

Abstract Background The shift of oral microbiota is a critical factor of radiation caries in head and neck cancer patients after the radiotherapy. However, the direct effects of irradiation on the genome and virulence of cariogenic bacteria are poorly described. Here we investigated the genomic mutations and virulence change of Streptococcus mutans (S. mutans), the major cariogenic bacteria, exposed to the therapeutic doses of X-rays. Results X-ray reduced the survival fraction of S. mutans and impacted its biofilm formation. We isolated a biofilm formation-deficient mutant #858 whose genome only possessed three synonymous mutations (c.2043 T > C, c.2100C > T, c.2109A > G) in gtfB gene. The “silent mutation” of c.2043 T > C in gtfB gene can cause the down-regulation of all of the gtfs genes’ expression and decrease the GtfB enzyme secretion without the effect on the growth due to the codon bias. #858 and synonymous point mutation strain gtfB2043 T>C, similar to the gtfB gene null mutant Δ gtfB, can significantly decrease the extracellular polysaccharide production, biofilm formation and cariogenic capabilities both in vitro and in vivo compared with wild type. Conclusion The direct exposure of X-ray radiation can affect the genome and virulence of oral bacteria even at therapeutic doses. The synonymous mutations of genome are negligent factors for gene expression and related protein translation due to the codon usage frequency.


2020 ◽  
Vol 2020 ◽  
pp. 1-13 ◽  
Author(s):  
Kaixin Xiong ◽  
Xuan Chen ◽  
Hantao Hu ◽  
Huihui Hou ◽  
Peng Gao ◽  
...  

Objective. To investigate the antibacterial effect of a novel antimicrobial peptide containing oral spray GERM CLEAN on Streptococcus mutans (S. mutans) in vitro and further explore the related mechanisms at phenotypic and transcriptional levels. Methods. The disk diffusion method was used to preliminarily appraise the antimicrobial effect of GERM CLEAN. The minimal inhibitory concentration (MIC) of GREM CLEAN towards S. mutans was determined by the broth dilution method. S. mutans virulence-related phenotypic assays including initial adhesive assay, pH drop, exopolysaccharides (EPS), and biofilm formation measurements and quantitative real-time PCR (qRT-PCR) were further applied to detect the inhibitory mechanisms of GREM CLEAN at 1/2MIC. Results. The diameter (10.18 ± 1.744 mm) of inhibition zones formed by GERM CLEAN preliminarily indicated its inhibitory effect on the major cariogenic bacteria S. mutans. The minimal inhibitory concentration of GERM CLEAN on S. mutans was 100% mass fraction (the stock solution). The study of the antibacterial mechanism showed that GERM CLEAN had a certain inhibitory effect on the initial adhesion, acid production, extracellular polysaccharides (EPS) production, and biofilm formation of S. mutans. GERM CLEAN disturbed S. mutans biofilm physiology mainly through destruction of biofilm architecture and suppression of bacterial growth. The results of qRT-PCR further confirmed that the expression levels of EPS and lactic acid generation genes including gtfB, gtfC, gtfD, and ldh were significantly repressed by treating with GERM CLEAN, and this was consistent with our phenotypic results. Conclusion. The novel antimicrobial peptide containing oral spray GERM CLEAN has an anti-Streptococcus mutans effect and the inhibitory property may be due to suppression of the virulence factors of S. mutans including adhesive, acidogenicity, EPS, and biofilm formation.


Sign in / Sign up

Export Citation Format

Share Document