Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 333
Filter
1.
J Appl Oral Sci ; 32: e20240047, 2024.
Article in English | MEDLINE | ID: mdl-38922243

ABSTRACT

OBJECTIVE: To assess the efficacy of Phyllanthus emblica extract in alleviating halitosis and reducing the inflammatory response to halitosis-related bacteria. METHODOLOGY: This investigation, using Phyllanthus emblica fruit extract (PE), involved four aspects. First, we evaluated the effect on growth and aggregation of halitosis-related bacteria, including Fusobacterium nucleatum, Porphyromonas gingivalis, and Solobacterium moorei, using a microdilution assay and scanning electron microscopy. Second, volatile sulfur compound (VSC) levels were measured on individuals with halitosis in randomized short-term (26 participants) and double-blind randomized long-term trials (18 participants in each group) after rinsing with PE for 3, 6, and 12 h, and 28 days. Third, we analyzed pro-inflammatory cytokine expression in TR146 cells using quantitative real-time PCR and enzyme-linked immunosorbent assays. Lastly, we assessed pro-inflammatory cytokine secretion and Toll-like receptor (TLR) 2 mRNA expression via the same experimental methods in a three-dimensional oral mucosal epithelial model (3D OMEM). RESULTS: PE extract dose-dependently inhibited the growth of F. nucleatum (50% inhibition concentration [IC50]=0.079%), P. gingivalis (IC50=0.65%), and S. moorei (IC50=0.07%) and effectively prevented bacterial aggregation. Furthermore, VSC contents decreased significantly at 3, 6, and 12 h after rinsing with 5% PE compared with those in the control. Long-term use of mouthwash containing 5% PE for 28 days led to a significant decrease in VSC contents. PE attenuated the F. nucleatum- or P. gingivalis-stimulated mRNA expression and protein release of interleukin (IL)-6 and IL-8 in TR146 cells. It also suppressed IL-8 and prostaglandin E2 secretion and TLR2 mRNA expression in F. nucleatum-induced OMEMs. CONCLUSION: Our findings support the use of PE in oral care products to alleviate halitosis and it may reduce inflammation.


Subject(s)
Cytokines , Enzyme-Linked Immunosorbent Assay , Fusobacterium nucleatum , Halitosis , Microscopy, Electron, Scanning , Phyllanthus emblica , Plant Extracts , Porphyromonas gingivalis , Real-Time Polymerase Chain Reaction , Phyllanthus emblica/chemistry , Halitosis/drug therapy , Halitosis/microbiology , Humans , Plant Extracts/pharmacology , Double-Blind Method , Fusobacterium nucleatum/drug effects , Porphyromonas gingivalis/drug effects , Female , Time Factors , Male , Treatment Outcome , Adult , Young Adult , Toll-Like Receptor 2/drug effects , Fruit/chemistry , Statistics, Nonparametric , Mouth Mucosa/drug effects , Mouth Mucosa/microbiology , Analysis of Variance , Sulfur Compounds/pharmacology , Sulfur Compounds/analysis
2.
Virulence ; 15(1): 2350904, 2024 12.
Article in English | MEDLINE | ID: mdl-38725098

ABSTRACT

Fusobacterium nucleatum (F. nucleatum) is closely correlated with tumorigenesis in colorectal cancer (CRC). We aimed to investigate the effects of host norepinephrine on the carcinogenicity of F. nucleatum in CRC and reveal the underlying mechanism. The results revealed that both norepinephrine and bacterial quorum sensing (QS) molecule auto-inducer-2 (AI-2) were positively associated with the progression of F. nucleatum related CRC (p < 0.01). In vitro studies, norepinephrine induced upregulation of QS-associated genes and promoted the virulence and proliferation of F. nucleatum. Moreover, chronic stress significantly increased the colon tumour burden of ApcMin/+ mice infected with F. nucleatum (p < 0.01), which was decreased by a catecholamine inhibitor (p < 0.001). Our findings suggest that stress-induced norepinephrine may promote the progression of F. nucleatum related CRC via bacterial QS signalling. These preliminary data provide a novel strategy for the management of pathogenic bacteria by targeting host hormones-bacterial QS inter-kingdom signalling.


Subject(s)
Colorectal Neoplasms , Fusobacterium nucleatum , Norepinephrine , Quorum Sensing , Signal Transduction , Quorum Sensing/drug effects , Fusobacterium nucleatum/pathogenicity , Fusobacterium nucleatum/drug effects , Fusobacterium nucleatum/physiology , Animals , Colorectal Neoplasms/microbiology , Norepinephrine/pharmacology , Mice , Humans , Disease Progression , Fusobacterium Infections/microbiology , Virulence , Homoserine/analogs & derivatives , Homoserine/metabolism , Mice, Inbred C57BL , Male , Lactones
3.
J Photochem Photobiol B ; 256: 112926, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38714001

ABSTRACT

Periodontitis, a chronic infectious disease leading to gingival atrophy and potential tooth loss through alveolar bone resorption, is closely linked to the oral microbiome. Fusobacterium nucleatum, known to facilitate late-stage bacterial colonization in the oral microbiome, plays a crucial role in the onset of periodontitis. Controlling F. nucleatum abundance is vital for preventing and treating periodontal disease. Photodynamic therapy combined with 5-aminolevulinic acid (ALA-PDT) has been reported to be bactericidal against Pseudomonas aeruginosa and Staphylococcus aureus. We aimed to investigate the bactericidal potential of ALA-PDT against F. nucleatum, which was evaluated by examining the impact of varying 5-ALA concentrations, culture time, and light intensity. After ALA-PDT treatment, DNA was extracted from interdental plaque samples collected from 10 volunteers and sequenced using the Illumina MiSeq platform. To further elucidate the bactericidal mechanism of ALA-PDT, porphyrins were extracted from F. nucleatum following cultivation with 5-ALA and subsequently analyzed using fluorescence spectra. ALA-PDT showed a significant bactericidal effect against F. nucleatum. Its bactericidal activity demonstrated a positive correlation with culture time and light intensity. Microbiota analysis revealed no significant alteration in α-diversity within the ALA-PDT group, although there was a noteworthy reduction in the proportion of the genus Fusobacterium. Furthermore, fluorescence spectral analysis indicated that F. nucleatum produced an excitable photosensitive substance following the addition of 5-ALA. Overall, if further studies confirm these results, this combined therapy could be an effective strategy for reducing the prevalence of periodontitis.


Subject(s)
Aminolevulinic Acid , Fusobacterium nucleatum , Periodontitis , Photochemotherapy , Photosensitizing Agents , Fusobacterium nucleatum/drug effects , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/therapeutic use , Humans , Periodontitis/microbiology , Periodontitis/drug therapy , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Adult , Male , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Female , Microbiota/drug effects
4.
Microbiol Spectr ; 12(7): e0342623, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38771061

ABSTRACT

Porphyromonas gingivalis has been associated with progression of periodontitis, characterized by inflammation and destruction of periodontal tissues. Here, we report that matcha, a product of Camellia sinensis, hampers the adherence and survival of P. gingivalis through multiple tactics. Matcha extract (ME) inhibited the growth not only of P. gingivalis but also of Prevotella nigrescens and Fusobacterium nucleatum, while it did not inhibit growth of nine species of oral streptococci and Aggregatibacter actinomycetemcomitans. ME-mediated P. gingivalis growth inhibition was characterized by both morphological and physiological changes at the bacterial envelope, which were accompanied by nano-particle formation and decreased membrane fluidity/permeability without loss of membrane integrity. ME also triggered autoaggregation of P. gingivalis in a major fimbriae (FimA)-dependent manner. In addition, adherence of P. gingivalis was dramatically inhibited by ME, irrespective of fimbriae. Furthermore, a structure-activity relationship study tested a series of catechins isolated from ME and identified the pyrogallol-type B-ring of catechins as essential for P. gingivalis growth inhibition. In a clinical study to assess the microbiological and therapeutic effects of matcha mouthwash in patients with periodontitis, the P. gingivalis number in saliva was significantly reduced by matcha mouthwash compared to the pre-intervention level. A tendency toward improvement in probing pocket depth was observed in the matcha group, although the difference was not statistically significant. Taken together, we present a proof of concept, based on the multimodal inhibitory effect of matcha against P. gingivalis, and that matcha may have clinical applicability for prevention and treatment of periodontitis. IMPORTANCE: Periodontitis, a multifactorial inflammatory disease of the oral cavity, results in alveolar bone destruction, and is a major cause of tooth loss of humans. In addition, emerging evidence has demonstrated associations between periodontitis and a wide range of other chronic inflammation-driven disorders, including diabetes mellitus, preterm birth, cardiovascular disease, aspiration pneumonia, rheumatoid arthritis, cognitive disorder, and cancer. In the present study, we report that matcha, a product of Camellia sinensis, hampers Porphyromonas gingivalis, a major periodontal pathobiont, in not only a series of in vitro experiments but also a pilot intervention clinical trial of patients with periodontitis, in which matcha mouthwash statistically significantly reduced the P. gingivalis number in saliva, as compared to the pre-intervention level. Taken together, we suggest that matcha may have clinical applicability for prevention and treatment of periodontitis.


Subject(s)
Anti-Bacterial Agents , Bacterial Adhesion , Periodontitis , Porphyromonas gingivalis , Porphyromonas gingivalis/drug effects , Porphyromonas gingivalis/growth & development , Porphyromonas gingivalis/physiology , Humans , Periodontitis/microbiology , Anti-Bacterial Agents/pharmacology , Bacterial Adhesion/drug effects , Plant Extracts/pharmacology , Plant Extracts/chemistry , Catechin/pharmacology , Fusobacterium nucleatum/drug effects , Fusobacterium nucleatum/growth & development , Fusobacterium nucleatum/physiology , Adult , Prevotella nigrescens/drug effects , Prevotella nigrescens/physiology , Female , Bacteroidaceae Infections/microbiology , Male , Aggregatibacter actinomycetemcomitans/drug effects , Aggregatibacter actinomycetemcomitans/growth & development , Aggregatibacter actinomycetemcomitans/physiology
5.
BMC Oral Health ; 24(1): 525, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702623

ABSTRACT

OBJECTIVE: To evaluate the antibacterial effectiveness of a combination of ε-poly-L-lysine (ε-PL), funme peptide (FP) as well as domiphen against oral pathogens, and assess the efficacy of a BOP® mouthwash supplemented with this combination in reducing halitosis and supragingival plaque in a clinical trial. MATERIALS AND METHODS: The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of the compound against Fusobacterium nucleatum, Porphyromonas gingivalis, Streptococcus mutans, and Aggregatibacter actinomycetemcomitans were determined by the gradient dilution method. Subsequently, the CCK-8 assay was used to detect the toxicity of mouthwash on human gingival fibroblastst, and the effectiveness in reducing halitosis and supragingival plaque of the mouthwash supplemented with the combination was analyzed by a randomized, double-blind, parallel-controlled clinical trial. RESULTS: The combination exhibited significant inhibitory effects on tested oral pathogens with the MIC < 1.56% (v/v) and the MBC < 3.13% (v/v), and the mouthwash containing this combination did not inhibit the viability of human gingival fibroblasts at the test concentrations. The clinical trial showed that the test group displayed notably lower volatile sulfur compounds (VSCs) at 0, 10, 24 h, and 7 d post-mouthwash (P < 0.05), compared with the baseline. After 7 days, the VSC levels of the and control groups were reduced by 50.27% and 32.12%, respectively, and notably cutting severe halitosis by 57.03% in the test group. Additionally, the Plaque Index (PLI) of the test and control group decreased by 54.55% and 8.38%, respectively, and there was a significant difference in PLI between the two groups after 7 days (P < 0.01). CONCLUSIONS: The combination of ε-PL, FP and domiphen demonstrated potent inhibitory and bactericidal effects against the tested oral pathogens, and the newly formulated mouthwash added with the combination exhibited anti-dental plaque and anti-halitosis properties in a clinical trial and was safe. TRIAL REGISTRATION: The randomized controlled clinical trial was registered on Chinese Clinical Trial Registry (No. ChiCTR2300073816, Date: 21/07/2023).


Subject(s)
Dental Plaque , Halitosis , Mouthwashes , Polylysine , Humans , Halitosis/prevention & control , Halitosis/drug therapy , Halitosis/microbiology , Mouthwashes/therapeutic use , Dental Plaque/microbiology , Dental Plaque/prevention & control , Double-Blind Method , Male , Female , Polylysine/therapeutic use , Adult , Microbial Sensitivity Tests , Young Adult , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Porphyromonas gingivalis/drug effects , Fusobacterium nucleatum/drug effects , Fibroblasts/drug effects , Peptides/therapeutic use , Peptides/pharmacology , Aggregatibacter actinomycetemcomitans/drug effects , Streptococcus mutans/drug effects
6.
ACS Nano ; 18(22): 14312-14326, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38767151

ABSTRACT

Periodontitis, a prevalent chronic inflammatory disease worldwide, is triggered by periodontopathogenic bacteria, resulting in the progressive destruction of periodontal tissue, particularly the alveolar bone. To effectively address periodontitis, this study proposed a nanoformulation known as CuS@MSN-SCS. This formulation involves coating citrate-grafted copper sulfide (CuS) nanoparticles with mesoporous silica (MSNs), followed by surface modification using amino groups and sulfated chitosan (SCS) through electrostatic interactions. The objective of this formulation is to achieve efficient bacteria removal by inducing ROS signaling pathways mediated by Cu2+ ions. Additionally, it aims to promote alveolar bone regeneration through Cu2+-induced pro-angiogenesis and SCS-mediated bone regeneration. As anticipated, by regulating the surface charges, the negatively charged CuS nanoparticles capped with sodium citrate were successfully coated with MSNs, and the subsequent introduction of amine groups using (3-aminopropyl)triethoxysilane was followed by the incorporation of SCS through electrostatic interactions, resulting in the formation of CuS@MSN-SCS. The developed nanoformulation was verified to not only significantly exacerbate the oxidative stress of Fusobacterium nucleatum, thereby suppressing bacteria growth and biofilm formation in vitro, but also effectively alleviate the inflammatory response and promote alveolar bone regeneration without evident biotoxicity in an in vivo rat periodontitis model. These findings contribute to the therapeutic effect on periodontitis. Overall, this study successfully developed a nanoformulation for combating bacteria and facilitating alveolar bone regeneration, demonstrating the promising potential for clinical treatment of periodontitis.


Subject(s)
Anti-Bacterial Agents , Bone Regeneration , Chitosan , Copper , Fusobacterium nucleatum , Nanoparticles , Periodontitis , Chitosan/chemistry , Chitosan/pharmacology , Periodontitis/drug therapy , Periodontitis/microbiology , Periodontitis/therapy , Periodontitis/pathology , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Bone Regeneration/drug effects , Rats , Copper/chemistry , Copper/pharmacology , Fusobacterium nucleatum/drug effects , Nanoparticles/chemistry , Rats, Sprague-Dawley , Male , Sulfates/chemistry , Sulfates/pharmacology , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacology , Microbial Sensitivity Tests
7.
ACS Infect Dis ; 10(5): 1612-1623, 2024 05 10.
Article in English | MEDLINE | ID: mdl-38597503

ABSTRACT

Fusobacterium nucleatum, a pathobiont inhabiting the oral cavity, contributes to opportunistic diseases, such as periodontal diseases and gastrointestinal cancers, which involve microbiota imbalance. Broad-spectrum antimicrobial agents, while effective against F. nucleatum infections, can exacerbate dysbiosis. This necessitates the discovery of more targeted narrow-spectrum antimicrobial agents. We therefore investigated the potential for the fusobacterial enoyl-ACP reductase II (ENR II) isoenzyme FnFabK (C4N14_ 04250) as a narrow-spectrum drug target. ENRs catalyze the rate-limiting step in the bacterial fatty acid synthesis pathway. Bioinformatics revealed that of the four distinct bacterial ENR isoforms, F. nucleatum specifically encodes FnFabK. Genetic studies revealed that fabK was indispensable for F. nucleatum growth, as the gene could not be deleted, and silencing of its mRNA inhibited growth under the test conditions. Remarkably, exogenous fatty acids failed to rescue growth inhibition caused by the silencing of fabK. Screening of synthetic phenylimidazole analogues of a known FabK inhibitor identified an inhibitor (i.e., 681) of FnFabK enzymatic activity and F. nucleatum growth, with an IC50 of 2.1 µM (1.0 µg/mL) and a MIC of 0.4 µg/mL, respectively. Exogenous fatty acids did not attenuate the activity of 681 against F. nucleatum. Furthermore, FnFabK was confirmed as the intracellular target of 681 based on the overexpression of FnFabK shifting MICs and 681-resistant mutants having amino acid substitutions in FnFabK or mutations in other genetic loci affecting fatty acid biosynthesis. 681 had minimal activity against a range of commensal flora, and it was less active against streptococci in physiologic fatty acids. Taken together, FnFabK is an essential enzyme that is amenable to drug targeting for the discovery and development of narrow-spectrum antimicrobial agents.


Subject(s)
Anti-Bacterial Agents , Fusobacterium nucleatum , Fusobacterium nucleatum/enzymology , Fusobacterium nucleatum/drug effects , Fusobacterium nucleatum/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Humans , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/genetics , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Microbial Sensitivity Tests , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/antagonists & inhibitors , Fatty Acids/chemistry , Fusobacterium Infections/microbiology , Fusobacterium Infections/drug therapy , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry
8.
Int J Mol Sci ; 25(8)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38673822

ABSTRACT

Complex microbial communities have been reported to be involved in endodontic infections. The microorganisms invade the dental pulp leading to pulpitis and initiating pulp inflammation. Fusobacterium nucleatum is a dominant bacterium implicated in both primary and secondary endodontic infections. Drugs targeting the molecular machinery of F. nucleatum will minimize pulp infection. LpxA and LpxD are early acyltransferases involved in the formation of lipid A, a major component of bacterial membranes. The identification of leads which exhibit preference towards successive enzymes in a single pathway can also prevent the development of bacterial resistance. A stringent screening strategy utilizing physicochemical and pharmacokinetic parameters along with a virtual screening approach identified two compounds, Lomefloxacin and Enoxacin, with good binding affinity towards the early acyltransferases LpxA and LpxD. Lomefloxacin and Enoxacin, members of the fluoroquinolone antibiotic class, exhibit wide-ranging activity against diverse bacterial strains. Nevertheless, their effectiveness in the context of endodontic treatment requires further investigation. This study explored the potential of Lomefloxacin and Enoxacin to manage endodontic infections via computational analysis. Moreover, the compounds identified herein serve as a foundation for devising novel combinatorial libraries with enhanced efficacy for endodontic therapeutic strategies.


Subject(s)
Anti-Bacterial Agents , Fusobacterium nucleatum , Lipopolysaccharides , Fusobacterium nucleatum/drug effects , Fusobacterium nucleatum/metabolism , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Lipopolysaccharides/metabolism , Molecular Docking Simulation , Computer Simulation , Fusobacterium Infections/drug therapy , Fusobacterium Infections/microbiology , Enoxacin/pharmacology , Bacterial Proteins/metabolism , Pulpitis/drug therapy , Pulpitis/metabolism , Pulpitis/microbiology
9.
J Mater Chem B ; 12(16): 3947-3958, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38586917

ABSTRACT

Colorectal cancer (CRC) occurs in the colorectum and ranks second in the global incidence of all cancers, accounting for one of the highest mortalities. Although the combination chemotherapy regimen of 5-fluorouracil (5-FU) and platinum(IV) oxaliplatin prodrug (OxPt) is an effective strategy for CRC treatment in clinical practice, chemotherapy resistance caused by tumor-resided Fusobacterium nucleatum (Fn) could result in treatment failure. To enhance the efficacy and improve the biocompatibility of combination chemotherapy, we developed an antibacterial-based nanodrug delivery system for Fn-associated CRC treatment. A tumor microenvironment-activated nanomedicine 5-FU-LA@PPL was constructed by the self-assembly of chemotherapeutic drug derivatives 5-FU-LA and polymeric drug carrier PPL. PPL is prepared by conjugating lauric acid (LA) and OxPt to hyperbranched polyglycidyl ether. In principle, LA is used to selectively combat Fn, inhibit autophagy in CRC cells, restore chemosensitivity of 5-FU as well as OxPt, and consequently enhance the combination chemotherapy effects for Fn-associated drug-resistant colorectal tumor. Both in vitro and in vivo studies exhibited that the tailored nanomedicine possessed efficient antibacterial and anti-tumor activities with improved biocompatibility and reduced non-specific toxicity. Hence, this novel anti-tumor strategy has great potential in the combination chemotherapy of CRC, which suggests a clinically relevant valuable option for bacteria-associated drug-resistant cancers.


Subject(s)
Antineoplastic Agents , Colorectal Neoplasms , Fluorouracil , Lauric Acids , Fluorouracil/pharmacology , Fluorouracil/chemistry , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Humans , Lauric Acids/chemistry , Lauric Acids/pharmacology , Animals , Mice , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Fusobacterium nucleatum/drug effects , Oxaliplatin/pharmacology , Oxaliplatin/chemistry , Drug Delivery Systems , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Prodrugs/chemistry , Prodrugs/pharmacology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Mice, Inbred BALB C , Particle Size , Drug Carriers/chemistry
10.
J Oral Biosci ; 66(2): 439-446, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38220090

ABSTRACT

OBJECTIVES: This study aimed to isolate antibacterial compounds active against periodontopathic bacteria from mulberry (Morus alba) leaves. METHODS: The acetone-soluble fraction of mulberry leaves was extracted from the oil layer by oil/water separation. The extract was purified using silica gel open-column chromatography. The minimum inhibitory concentration (MIC) of the crude extract or purified fractions against Porphyromonas gingivalis was measured at each step. RESULTS: The MIC of the crude extract against P. gingivalis was 62.5-125 µg/mL. The fractions showing activity against P. gingivalis were designated Cf K and Cf P. The MICs of Cf K against P. gingivalis, Fusobacterium nucleatum, Prevotella intermedia, and Streptococcus mutans were 6.25 µg/mL, 25 µg/mL, 12.5 µg/mL, and 12.5 µg/mL, respectively. In contrast, the MICs of Cf P against P. gingivalis, F. nucleatum, P. intermedia, and S. mutans were 25.0 µg/mL, >50 µg/mL, 50 µg/mL, and 12.5-25.0 µg/mL, respectively. CONCLUSIONS: Mulberry leaves contain antibacterial components against periodontopathic bacteria such as P. gingivalis, F. nucleatum, and P. intermedia.


Subject(s)
Anti-Bacterial Agents , Fusobacterium nucleatum , Microbial Sensitivity Tests , Morus , Plant Extracts , Plant Leaves , Porphyromonas gingivalis , Streptococcus mutans , Morus/chemistry , Plant Extracts/pharmacology , Plant Extracts/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Porphyromonas gingivalis/drug effects , Plant Leaves/chemistry , Fusobacterium nucleatum/drug effects , Streptococcus mutans/drug effects , Prevotella intermedia/drug effects
11.
ACS Appl Mater Interfaces ; 14(1): 390-403, 2022 Jan 12.
Article in English | MEDLINE | ID: mdl-34935355

ABSTRACT

Silver-based nano-antibiotics are rapidly developing as promising alternatives to conventional antibiotics. Ideally, to remain potent against a wide range of drug-resistant and anaerobic bacteria, silver-based nano-antibiotics should easily penetrate through the bacterial cell walls and actively release silver ions. In this study, highly monodispersed, ultrasmall (<3 nm), polycationic silver nanoclusters (pAgNCs) are designed and synthesized for the elimination of a range of common Gram-negative and Gram-positive pathogens and their corresponding established and matured biofilms, including those composed of multiple species. The pAgNCs also show greatly enhanced antibacterial efficacy against anaerobic bacteria such as Fusobacterium nucleatum and Streptococcus sanguinis. These results demonstrate that the cationic nature facilitates better penetration to the bacterial cell membrane while the presence of a high percentage (>50%) of silver ions (i.e., Ag+ nanoreservoirs) on the cluster surface maintains their efficiency in both aerobic and anaerobic conditions. Significantly, the pAgNCs showed a strong capacity to significantly delay the development of bacterial resistance when compared to similar-sized negatively charged silver nanoparticles or conventional antibiotics. This study demonstrates a novel design strategy that can lay the foundation for the development of future highly potent nano-antibiotics effective against a broad spectrum of pathogens and biofilms needed in many everyday life applications and industries.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biocompatible Materials/pharmacology , Nanoparticles/chemistry , Polyelectrolytes/pharmacology , Silver/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Biocompatible Materials/chemical synthesis , Biocompatible Materials/chemistry , Biofilms/drug effects , Fusobacterium nucleatum/drug effects , Ions/chemistry , Ions/pharmacology , Materials Testing , Microbial Sensitivity Tests , Particle Size , Polyelectrolytes/chemistry , Silver/chemistry , Streptococcus sanguis/drug effects
12.
J Mater Chem B ; 9(46): 9594-9605, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34783814

ABSTRACT

Fusobacterium nucleatum caused drug-resistant around tumor sites often leads to the failure of chemotherapy during colorectal cancer (CRC) treatment. Multifunctional cationic quaternary ammonium materials have been widely used as broad-spectrum antibacterial agents in antibacterial and anticancer fields. Herein, we design a smart supramolecular quaternary ammonium nanoparticle, namely quaternary ammonium PAMAM-AZO@CP[5]A (Q-P-A@CP[5]A), consisting of azobenzene (AZO)-conjugated dendritic cationic quaternary ammonium polyamidoamine (PAMAM) as the core and carboxylatopillar[5]arene (CP[5]A)-based switch, for antibacterial and anti-CRC therapies. The quaternary ammonium-PAMAM-AZO (Q-P-A) core endows the supramolecular system with enhanced antibacterial and anticancer properties. -N+CH3 groups on the surface of Q-P-A are accommodated in the CP[5]A cavity under normal conditions, which significantly improves the biocompatibility of Q-P-A@CP[5]A. Meanwhile, the CP[5]A host can be detached from -N+CH3 groups under pathological conditions, achieving efficient antibacterial and antitumor therapies. Furthermore, azoreductase in the tumor site can break the -NN- bonds of AZO in Q-P-A@CP[5]A, leading to the morphology recovery of supramolecular nanoparticles and CRC therapy through inducing cell membrane rupture. Both in vitro and in vivo experiments demonstrate that Q-P-A@CP[5]A possesses good biocompatibility, excellent antibacterial effect, and CRC treatment capability with negligible side effects. This supramolecular quaternary ammonium system provides an effective treatment method to overcome chemotherapy-resistant cancer caused by bacteria.


Subject(s)
Fusobacterium nucleatum/drug effects , Nanoparticles/chemistry , Polyamines/chemistry , Quaternary Ammonium Compounds/chemistry , Animals , Anti-Bacterial Agents , Antifungal Agents , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis , Azo Compounds/chemistry , Cell Survival , Drug Design , Drug Resistance, Fungal , Drug Resistance, Neoplasm , HT29 Cells , Humans , Male , Mice , Mice, Nude , Models, Molecular , Molecular Structure , Neoplasms, Experimental , Xenograft Model Antitumor Assays
13.
ACS Appl Mater Interfaces ; 13(42): 49642-49657, 2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34637255

ABSTRACT

Periodontitis compromises the integrity and function of tooth-supporting structures. Although therapeutic approaches have been offered, predictable regeneration of periodontal tissues remains intangible, particularly in anatomically complex defects. In this work, personalized and defect-specific antibiotic-laden polymeric scaffolds containing metronidazole (MET), tetracycline (TCH), or their combination (MET/TCH) were created via electrospinning. An initial screening of the synthesized fibers comprising chemo-morphological analyses, cytocompatibility assessment, and antimicrobial validation against periodontopathogens was accomplished to determine the cell-friendly and anti-infective nature of the scaffolds. According to the cytocompatibility and antimicrobial data, the 1:3 MET/TCH formulation was used to obtain three-dimensional defect-specific scaffolds to treat periodontally compromised three-wall osseous defects in rats. Inflammatory cell response and new bone formation were assessed by histology. Micro-computerized tomography was performed to assess bone loss in the furcation area at 2 and 6 weeks post implantation. Chemo-morphological and cell compatibility analyses confirmed the synthesis of cytocompatible antibiotic-laden fibers with antimicrobial action. Importantly, the 1:3 MET/TCH defect-specific scaffolds led to increased new bone formation, lower bone loss, and reduced inflammatory response when compared to antibiotic-free scaffolds. Altogether, our results suggest that the fabrication of defect-specific antibiotic-laden scaffolds holds great potential toward the development of personalized (i.e., patient-specific medication) scaffolds to ablate infection while affording regenerative properties.


Subject(s)
Anti-Bacterial Agents/pharmacology , Metronidazole/pharmacology , Periodontitis/drug therapy , Tetracycline/pharmacology , Tissue Scaffolds/chemistry , Anti-Bacterial Agents/chemistry , Bone Regeneration/drug effects , Fusobacterium nucleatum/drug effects , Materials Testing , Metronidazole/chemistry , Microbial Sensitivity Tests , Particle Size , Periodontitis/microbiology , Periodontitis/pathology , Porphyromonas gingivalis/drug effects , Prevotella intermedia/drug effects , Tetracycline/chemistry
14.
Food Funct ; 12(21): 10470-10483, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34554173

ABSTRACT

Cranberries are widely recognized as a functional food that can promote oral health. However, the high concentration of organic acids in cranberry juice can cause tooth enamel erosion. Electrodialysis with bipolar membrane (EDBM) is a process used for the deacidification of cranberry juice. The present study investigated whether the removal of organic acids (0%, 19%, 42%, 60%, and 79%) from cranberry juice by EDBM affects its antibacterial activity against major periodontopathogens as well as its anti-inflammatory properties in an oral epithelial cell model. A deacidification rate ≥60% attenuated the bactericidal effect against planktonic and biofilm-embedded Aggregatibacter actinomycetemcomitans but had no impact on Porphyromonas gingivalis and Fusobacterium nucleatum. Cranberry juice increased the adherence of A. actinomycetemcomitans and P. gingivalis to oral epithelial cells, but reduced the adherence of F. nucleatum by half regardless of the deacidification rate. F. nucleatum produced more hydrogen sulfide when it was exposed to deacidified cranberry juice with a deacidification rate ≥42% compared to the raw beverage. Interestingly, the removal of organic acids from cranberry juice lowered the cytotoxicity of the beverage for oral epithelial cells. Deacidification attenuated the anti-inflammatory effect of cranberry juice in an in vitro oral epithelial cell model. The secretion of IL-6 by lipopolysaccharide (LPS)-stimulated oral epithelial cells exposed to cranberry juice increased proportionally with the deacidification rate. No such effect was observed with respect to the production of IL-8. This study provided evidence that organic acids, just like phenolic compounds, might contribute to the health benefits of cranberry juice against periodontitis.


Subject(s)
Aggregatibacter actinomycetemcomitans/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Fusobacterium nucleatum/drug effects , Plant Extracts/pharmacology , Porphyromonas gingivalis/drug effects , Vaccinium macrocarpon/chemistry , Acids/chemistry , Anti-Bacterial Agents/chemistry , Anti-Inflammatory Agents/chemistry , Biofilms , Cells, Cultured , Electrochemical Techniques/methods , Epithelial Cells/drug effects , Fruit and Vegetable Juices , Plant Extracts/chemistry
15.
J Infect Dev Ctries ; 15(7): 1027-1031, 2021 07 31.
Article in English | MEDLINE | ID: mdl-34343129

ABSTRACT

INTRODUCTION: Propolis is a natural composite balsam. In the past decade, propolis has been extensively investigated as an adjuvant for the treatment of periodontitis. This study aimed to investigate antimicrobial activities of propolis solutions and plant essential oils against some oral cariogenic (Streptococcus mutans, Streptococcus mitis, Streptococcus sanguis, Lactobacillus acidophilus) and periodontopathic bacteria (Actinomyces odontolyticus, Eikenella corrodens, Fusobacterium nucleatum). METHODOLOGY: Determination of the minimum inhibitory concentration (MIC): The antimicrobial activity of propolis and essential oils was investigated by the agar dilution method. Serial dilutions of essential oils were prepared in plates, and the assay plates were estimated to contain 100, 50, 25 and 12.5 µg/mL of active essential oils. Dilutions for propolis were 50, 25, 12.5 and 6.3 µg/mL of active propolis solutions. RESULTS: Propolis solutions dissolved in benzene, diethyl ether and methyl chloride, demonstrated equal effectiveness against all investigated oral bacteria (MIC=12.5 µg/mL). Propolis solution dissolved in acetone displayed MIC of 6.3 µg/mL only for Lactobacillus acidophilus. At the MIC of 12.5 µg/mL, essential oils of Salvia officinalis and Satureja kitaibelii were effective against Streptococcus mutans and Porphyromonas gingivalis, respectively. For the latter, the MIC value of Salvia officinalis was twice higher. CONCLUSIONS: The results indicate that propolis and plant essential oils appear to be a promising source of antimicrobial agents that may prevent dental caries and other oral infectious diseases.


Subject(s)
Anti-Infective Agents/pharmacology , Lactobacillus acidophilus/drug effects , Oils, Volatile/pharmacology , Porphyromonas gingivalis/drug effects , Propolis/pharmacology , Streptococcus mutans/drug effects , Actinomyces/drug effects , Eikenella corrodens/drug effects , Fusobacterium nucleatum/drug effects , Humans , Microbial Sensitivity Tests , Mouth/microbiology , Prospective Studies , Salvia officinalis/chemistry , Satureja/chemistry , Streptococcus mitis/drug effects , Streptococcus sanguis/drug effects
16.
APMIS ; 129(11): 626-630, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34418158

ABSTRACT

Species (spp.) belonging to the genus Fusobacterium are anaerobic commensals colonizing the upper respiratory tract, the gastrointestinal tract, and the genitals. Infections with Fusobacterium spp. have been reported at many anatomical sites, including pneumonias and pleural empyemas; however, there are very few published cases of Fusobacterium spp. causing spondylodiscitis or fistulas. Bone infections with Fusobacterium can spread directly to surrounding muscular tissue or by hematogenous transmission to any other tissue including pleurae and lungs. Similarly, pleural infections can spread Fusobacterium spp. to any other tissue including fistulas and bone. Concomitant pleural empyema and spondylodiscitis are rare; however, there are a few published cases with concomitant disease, although none caused by Fusobacterium spp. A 77-year-old female patient was assessed using computed tomography (CT) scanning of the thorax and abdomen, as well as analyses of fluid drained from the region affected by the pleural empyema. A diagnosis of Fusobacterium empyema, fistula, bacteremia, and spondylodiscitis was made, and the patient's condition improved significantly after drainage of the pleural empyema and relevant long-term antibiotic treatment. We describe the first confirmed case with concomitant infection with Fusobacterium nucleatum as spondylodiscitis and pleural empyema connected by a fistula.


Subject(s)
Discitis/etiology , Empyema, Pleural/etiology , Fistula/etiology , Fusobacterium Infections/complications , Fusobacterium nucleatum/pathogenicity , Aged , Anti-Bacterial Agents/therapeutic use , Bacteremia/diagnosis , Bacteremia/drug therapy , Bacteremia/etiology , Discitis/diagnosis , Discitis/drug therapy , Empyema, Pleural/diagnosis , Empyema, Pleural/drug therapy , Female , Fistula/diagnosis , Fistula/drug therapy , Fusobacterium Infections/diagnosis , Fusobacterium Infections/drug therapy , Fusobacterium nucleatum/drug effects , Humans , Pleura/diagnostic imaging , Pleura/microbiology , Treatment Outcome
17.
Probiotics Antimicrob Proteins ; 13(6): 1808-1819, 2021 12.
Article in English | MEDLINE | ID: mdl-34402021

ABSTRACT

This study evaluated the cytocompatibility and antimicrobial/antibiofilm effects of epigallocatechin-3-gallate (EGCG) associated with peptide LL-37 and its analogue KR-12-a5 against oral pathogens. The effect of the compounds on metabolism of fibroblasts was evaluated by methyltetrazolium assays. Antimicrobial activity of the compounds was evaluated on Streptococcus mutans, Enterococcus faecalis, Actinomyces israelii, and Fusobacterium nucleatum under planktonic conditions, on single- and dual-species biofilms and E. faecalis biofilms in dentinal tubules and analyzed by bacterial counts and confocal microscopy. Data were statistically analyzed considering p < 0.05. EGCG and peptide combinations were not toxic to fibroblasts. KR-12-a5 showed synergistic or addictive effects with EGCG and LL-37 against all bacteria tested. However, EGCG associated with KR-12-a5 demonstrated the highest bactericidal activity on all bacteria tested, at lower concentrations. In single-species biofilms, EGCG + KR-12-a5 eliminated S. mutans and A. israelii and reduced E. faecalis and F. nucleatum counts around 5 log CFU/mL. EGCG + KR-12-a5 reduced E. faecalis (-3.93 log CFU/mL) and eliminated S. mutans in dual-species biofilms. No growth of E. faecalis and significant reduction in A. israelii (-6.24 log CFU/mL) and F. nucleatum (-4.62 log CFU/mL) counts were detected in dual-species biofilms. The combination of EGCG and KR-12-a5 led to 88% of E. faecalis dead cells inside dentin tubules. The association of EGCG and KR-12-a5 was cytocompatible and promoted synergistic effect against biofilms of bacteria associated with endodontic infections.


Subject(s)
Anti-Infective Agents , Biofilms/drug effects , Catechin/analogs & derivatives , Peptides/pharmacology , Actinomyces/drug effects , Anti-Infective Agents/pharmacology , Catechin/pharmacology , Enterococcus faecalis/drug effects , Fusobacterium nucleatum/drug effects , Streptococcus mutans/drug effects
18.
Molecules ; 26(16)2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34443332

ABSTRACT

Peptoids (oligo N-substituted glycines) are peptide analogues, which can be designed to mimic host antimicrobial peptides, with the advantage that they are resistant to proteolytic degradation. Few studies on the antimicrobial efficacy of peptoids have focused on Gram negative anaerobic microbes associated with clinical infections, which are commonly recalcitrant to antibiotic treatment. We therefore studied the cytotoxicity and antibiofilm activity of a family of peptoids against the Gram negative obligate anaerobe Fusobacterium nucleatum, which is associated with infections in the oral cavity. Two peptoids, peptoid 4 (NaeNpheNphe)4 and peptoid 9 (NahNspeNspe)3 were shown to be efficacious against F. nucleatum biofilms at a concentration of 1 µM. At this concentration, peptoids 4 and 9 were not cytotoxic to human erythrocytes or primary human gingival fibroblast cells. Peptoids 4 and 9 therefore have merit as future therapeutics for the treatment of oral infections.


Subject(s)
Biofilms/drug effects , Fusobacterium nucleatum/drug effects , Fusobacterium nucleatum/physiology , Peptoids/pharmacology , Biofilms/growth & development , Drug Resistance, Bacterial/drug effects
19.
Anaerobe ; 71: 102420, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34314865

ABSTRACT

A 42-year-old man was referred to the Department of Orthopedic Surgery with pain over his right greater trochanter and signs of systemic infection. CT showed an enhanced mass in his gluteus maximus as well as gas in the biceps femoris over the underlying hip joint. Tissue biopsy yielded Fusobacterium nucleatum and Actinomyces turicensis. The patient was successfully treated for 6 weeks with amoxicillin/clavulanic acid 875mg/125mg and metronidazole 500mg.


Subject(s)
Actinomycetaceae/isolation & purification , Actinomycetales Infections/microbiology , Bacteremia/microbiology , COVID-19/immunology , Fusobacterium Infections/microbiology , Fusobacterium nucleatum/isolation & purification , Hip/microbiology , Abscess/drug therapy , Abscess/microbiology , Actinomycetaceae/drug effects , Actinomycetaceae/genetics , Actinomycetales Infections/drug therapy , Adult , Anti-Bacterial Agents/therapeutic use , COVID-19/virology , Fusobacterium Infections/drug therapy , Fusobacterium nucleatum/drug effects , Fusobacterium nucleatum/genetics , Humans , Immunocompromised Host , Male , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification
20.
PLoS One ; 16(5): e0252029, 2021.
Article in English | MEDLINE | ID: mdl-34029354

ABSTRACT

Fusobacterium nucleatum is associated with many conditions and diseases, including periodontal diseases that affect tooth-supporting tissues. The aim of the present study was to investigate the effects of a cocoa extract (Theobroma cacao L.) on F. nucleatum with respect to growth, biofilm formation, adherence, and hydrogen sulfide (H2S) production. The anti-inflammatory properties and the effect on epithelial barrier function of the cocoa extract were also assessed. The cocoa extract, whose major phenolic compound is epicatechin, dose-dependently inhibited the growth, biofilm formation, adherence properties (basement membrane matrix, oral epithelial cells), and H2S production of F. nucleatum. It also decreased IL-6 and IL-8 production by F. nucleatum-stimulated oral epithelial cells and inhibited F. nucleatum-induced NF-κB activation in monocytes. Lastly, the cocoa extract enhanced the barrier function of an oral epithelial model by increasing the transepithelial electrical resistance. We provide evidence that the beneficial properties of an epicatechin-rich cocoa extract may be useful for preventing and/or treating periodontal diseases.


Subject(s)
Cacao/chemistry , Fusobacterium nucleatum/drug effects , Periodontal Diseases/drug therapy , Phenols/pharmacology , Biofilms/drug effects , Catechin/chemistry , Catechin/pharmacology , Cell Adhesion/drug effects , Epithelial Cells/drug effects , Fusobacterium nucleatum/pathogenicity , Gene Expression Regulation/drug effects , Humans , Hydrogen Sulfide/metabolism , Interleukin-6/genetics , Interleukin-8/genetics , Monocytes/drug effects , Periodontal Diseases/microbiology , Phenols/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...