Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Int J Mol Sci ; 22(24)2021 Dec 19.
Article in English | MEDLINE | ID: mdl-34948413

ABSTRACT

Proton pump inhibitors (PPI) may improve symptoms in functional dyspepsia (FD) through duodenal eosinophil-reducing effects. However, the contribution of the microbiome to FD symptoms and its interaction with PPI remains elusive. Aseptic duodenal brushings and biopsies were performed before and after PPI intake (4 weeks Pantoprazole 40 mg daily, FD-starters and controls) or withdrawal (2 months, FD-stoppers) for 16S-rRNA sequencing. Between- and within-group changes in genera or diversity and associations with symptoms or duodenal factors were analyzed. In total, 30 controls, 28 FD-starters and 19 FD-stoppers were followed. Mucus-associated Porphyromonas was lower in FD-starters vs. controls and correlated with symptoms in FD and duodenal eosinophils in both groups, while Streptococcus correlated with eosinophils in controls. Although clinical and eosinophil-reducing effects of PPI therapy were unrelated to microbiota changes in FD-starters, increased Streptococcus was associated with duodenal PPI effects in controls and remained higher despite withdrawal of long-term PPI therapy in FD-stoppers. Thus, duodenal microbiome analysis demonstrated differential mucus-associated genera, with a potential role of Porphyromonas in FD pathophysiology. While beneficial effects of short-term PPI therapy were not associated with microbial changes in FD-starters, increased Streptococcus and its association with PPIeffects in controls suggest a role for duodenal dysbiosis after long-term PPI therapy.


Subject(s)
Duodenum/microbiology , Dysbiosis/chemically induced , Dyspepsia/drug therapy , Proton Pump Inhibitors/therapeutic use , Adult , Duodenum/drug effects , Dysbiosis/microbiology , Dyspepsia/microbiology , Female , Gastrointestinal Microbiome/drug effects , Humans , Male , Middle Aged , Porphyromonas/drug effects , Proton Pump Inhibitors/adverse effects , Young Adult
2.
Anaerobe ; 71: 102442, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34481990

ABSTRACT

Porphyromonas species are Gram-negative anaerobic bacilli mainly involved in human periodontal diseases. We report an uncommon case of bacteremia due to P. asaccharolytica in a patient with necrotizing fasciitis. A 52-year-old woman with a history of diabetes mellitus was admitted for an extensive necrotizing lesion on the left lower limb. After she developed septic shock, two sets of blood cultures were taken. Anaerobic bottles yielded a pure culture of a microorganism initially identified as P. uenonis by MALDI-TOF MS but with a low log score, and a gene sequencing technique was therefore applied, identifying the isolate as P. asaccharolytica. Only resistance to penicillin and clindamycin was documented. Treatment with meropenem was administered, and the patient was discharged following her recovery.


Subject(s)
Bacteremia/microbiology , Fasciitis, Necrotizing/microbiology , Porphyromonas/physiology , Anti-Bacterial Agents/therapeutic use , Bacteremia/drug therapy , Fasciitis, Necrotizing/drug therapy , Female , Humans , Middle Aged , Porphyromonas/drug effects , Porphyromonas/genetics , Porphyromonas/isolation & purification
3.
Carbohydr Polym ; 260: 117777, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33712133

ABSTRACT

The combination of alginate, hyaluronic acid and multivalent ions have been reported to form alginate-hyaluronic acid ionic-crosslinking hydrogels for biomedical applications. However, injectable alginate-hyaluronic acid ionic-crosslinking hydrogels with satisfactory shear-thinning property have rarely been reported. In this study, we successfully developed an ionic-crosslinked alginate-hyaluronic acid hydrogel by simple assembly of alginate-hyaluronic acid mixture and Fe3+ complex. This hydrogel could fully recover within seconds after damaged, while displayed shear thinning behavior and good injectability which were contributed by the reversible and dynamic metal-ligand interactions formed via ferric ions and carboxyl groups of the polymers. Moreover, the local degradation of this hydrogel giving the hydrogel sustained ferric ions release property, of which led to potential long-term antibacterial activities against multiple types of bacteria including gram-negative Escherichia coli and gram-positive Staphylococcus aureus, as well as representative oral pathogenic bacteria Streptococcus mutans and Porphyromonas gingivalis.


Subject(s)
Alginates/chemistry , Anti-Infective Agents/chemistry , Ferric Compounds/chemistry , Hyaluronic Acid/chemistry , Hydrogels/chemistry , Animals , Anti-Infective Agents/pharmacology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Line , Cell Survival/drug effects , Escherichia coli/drug effects , Female , Ferric Compounds/metabolism , Humans , Hydrogels/pharmacology , Mice , Mice, Inbred BALB C , Porphyromonas/drug effects , Rheology , Skin/drug effects , Skin/pathology , Staphylococcus aureus/drug effects , Streptococcus mutans/drug effects
4.
J Appl Oral Sci ; 28: e20190519, 2020.
Article in English | MEDLINE | ID: mdl-32348444

ABSTRACT

Natural products have emerged as a rich source of bioactive compounds for adjunctive treatments of many infectious and inflammatory conditions, including periodontitis. Among the monoterpenes with significant biological properties, there is the perillyl alcohol (POH), which can be found in several essential oils and has shown immunomodulatory properties in recent studies, which may be interesting in the treatment of non-neoplastic inflammatory disorders. Objective To determine the antibacterial and immune modulatory activities of the POH. Methodology The minimum inhibitory concentration (MIC) and the minimum bactericidal concentration (MBC) of the POH for two significant Gram-negative periodontal pathogens were determined by macrodilution and subculture, respectively. Cell proliferation and cytotoxicity in RAW 264.7 macrophages were determined by Trypan Blue and mitochondrial enzymatic activity assay. The modulation of reactive oxygen species (ROS) was analyzed by flow cytometry and expression of TNF and arginase-1 by real-time PCR. Results The POH was effective against P. gingivalis (ATCC 33277) and F. nucleatum (ATCC 25586) with MIC= MBC=1600 µM. No cytotoxicity up to 100 µM was observed on macrophages. The cell proliferation was inhibited from 48 hours at 100 µM (p<0.05) and 250 µM (p<0.01). The POH increased ROS production at both 10 µM and 100 µM (p<0.05) in unstimulated cells. The PMA-induced ROS production was not affected by POH, whereas 100 µM significantly reduced lipopolysaccharide-induced (LPS-induced) ROS. The expression of TNF was not affected by POH in unstimulated cells or in cells polarized to M1 phenotype, whereas both concentrations of POH reduced (p<0.05) the expression of arginase-1 in M2-polarized macrophages. Conclusion The POH has antibacterial activity against periodontal pathogens and reduced proliferation of murine macrophages without significant cytotoxicity at concentrations up to 100 µM. In addition, the POH reduced the LPS-induced ROS and the expression of arginase-1 in M2-polarized macrophages.


Subject(s)
Anti-Bacterial Agents/pharmacology , Fusobacterium nucleatum/drug effects , Macrophages/drug effects , Monoterpenes/pharmacology , Porphyromonas/drug effects , Reactive Oxygen Species/analysis , Animals , Arginase/analysis , Biological Products/pharmacology , Cell Proliferation/drug effects , Flow Cytometry , Fusobacterium nucleatum/growth & development , Gene Expression , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , Microbial Sensitivity Tests , Porphyromonas/growth & development , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Time Factors , Tumor Necrosis Factor-alpha/analysis
5.
Sci Rep ; 10(1): 3113, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32080231

ABSTRACT

Porphyromonas gulae is a major periodontal pathogen in dogs, which can be transmitted to their owners. A major virulence factor of P. gulae consists of a 41-kDa filamentous appendage (FimA) on the cell surface, which is classified into three genotypes: A, B, and C. Thus far, inhibition of periodontal disease in dogs remains difficult. The present study assessed the inhibitory effects of a combination of clindamycin and interferon alpha (IFN-α) formulation against P. gulae and periodontal disease. Growth of P. gulae was significantly inhibited by clindamycin; this inhibition had a greater effect on type C P. gulae than on type A and B isolates. In contrast, the IFN-α formulation inhibited the expression of IL-1ß and COX-2 elicited by type A and B isolates, but not that elicited by type C isolates. Furthermore, periodontal recovery was promoted by the administration of both clindamycin and IFN-α formulation to dogs undergoing periodontal treatment; moreover, this combined treatment reduced the number of FimA genotypes in oral specimens from treated dogs. These results suggest that a combination of clindamycin and IFN-α formulation inhibit P. gulae virulence and thus may be effective for the prevention of periodontal disease induced by P. gulae.


Subject(s)
Clindamycin/administration & dosage , Interferon-alpha/administration & dosage , Periodontal Diseases/drug therapy , Periodontal Diseases/veterinary , Porphyromonas/drug effects , Animals , Bacteroidaceae Infections/drug therapy , Bacteroidaceae Infections/veterinary , Cell Line , Cytokines/metabolism , Dogs , Drug Design , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Female , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Genotype , Gingiva/drug effects , Gingiva/microbiology , Humans , Male , Virulence , Virulence Factors/metabolism
6.
Pharmacol Res Perspect ; 8(1): e00562, 2020 02.
Article in English | MEDLINE | ID: mdl-31999052

ABSTRACT

COR388, a small-molecule lysine-gingipain inhibitor, is currently being investigated in a Phase 2/3 clinical trial for Alzheimer's disease (AD) with exploratory endpoints in periodontal disease. Gingipains are produced by two species of bacteria, Porphyromonas gingivalis and Porphyromonas gulae, typically associated with periodontal disease and systemic infections in humans and dogs, respectively. P. gulae infection in dogs is associated with periodontal disease, which provides a physiologically relevant model to investigate the pharmacology of COR388. In the current study, aged dogs with a natural oral infection of P. gulae and periodontal disease were treated with COR388 by oral administration for up to 90 days to assess lysine-gingipain target engagement and reduction of bacterial load and downstream pathology. In a 28-day dose-response study, COR388 inhibited the lysine-gingipain target and reduced P. gulae load in saliva, buccal cells, and gingival crevicular fluid. The lowest effective dose was continued for 90 days and was efficacious in continuous reduction of bacterial load and downstream periodontal disease pathology. In a separate histology study, dog brain tissue showed evidence of P. gulae DNA and neuronal lysine-gingipain, demonstrating that P. gulae infection is systemic and spreads beyond its oral reservoir, similar to recent observations of P. gingivalis in humans. Together, the pharmacokinetics and pharmacodynamics of COR388 lysine-gingipain inhibition, along with reduction of bacterial load and periodontal disease in naturally occurring P. gulae infection in the dog, support the use of COR388 in targeting lysine-gingipain and eliminating P. gingivalis infection in humans.


Subject(s)
Bacteroidaceae Infections/drug therapy , Dog Diseases/microbiology , Gingipain Cysteine Endopeptidases/antagonists & inhibitors , Organic Chemicals/administration & dosage , Periodontal Diseases/drug therapy , Porphyromonas/enzymology , Small Molecule Libraries/administration & dosage , Administration, Oral , Aging/blood , Animals , Bacterial Load , Bacterial Proteins/antagonists & inhibitors , Bacteroidaceae Infections/veterinary , Brain/drug effects , Brain/microbiology , Dog Diseases/drug therapy , Dogs , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gingival Crevicular Fluid/drug effects , Gingival Crevicular Fluid/microbiology , Organic Chemicals/chemistry , Organic Chemicals/pharmacology , Periodontal Diseases/veterinary , Porphyromonas/drug effects , Porphyromonas/pathogenicity , Saliva/drug effects , Saliva/microbiology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
7.
J. appl. oral sci ; 28: e20190519, 2020. tab, graf
Article in English | LILACS, BBO - Dentistry | ID: biblio-1101254

ABSTRACT

Abstract Natural products have emerged as a rich source of bioactive compounds for adjunctive treatments of many infectious and inflammatory conditions, including periodontitis. Among the monoterpenes with significant biological properties, there is the perillyl alcohol (POH), which can be found in several essential oils and has shown immunomodulatory properties in recent studies, which may be interesting in the treatment of non-neoplastic inflammatory disorders. Objective To determine the antibacterial and immune modulatory activities of the POH. Methodology The minimum inhibitory concentration (MIC) and the minimum bactericidal concentration (MBC) of the POH for two significant Gram-negative periodontal pathogens were determined by macrodilution and subculture, respectively. Cell proliferation and cytotoxicity in RAW 264.7 macrophages were determined by Trypan Blue and mitochondrial enzymatic activity assay. The modulation of reactive oxygen species (ROS) was analyzed by flow cytometry and expression of TNF and arginase-1 by real-time PCR. Results The POH was effective against P. gingivalis (ATCC 33277) and F. nucleatum (ATCC 25586) with MIC= MBC=1600 μM. No cytotoxicity up to 100 µM was observed on macrophages. The cell proliferation was inhibited from 48 hours at 100 μM (p<0.05) and 250 μM (p<0.01). The POH increased ROS production at both 10 μM and 100 μM (p<0.05) in unstimulated cells. The PMA-induced ROS production was not affected by POH, whereas 100 μM significantly reduced lipopolysaccharide-induced (LPS-induced) ROS. The expression of TNF was not affected by POH in unstimulated cells or in cells polarized to M1 phenotype, whereas both concentrations of POH reduced (p<0.05) the expression of arginase-1 in M2-polarized macrophages. Conclusion The POH has antibacterial activity against periodontal pathogens and reduced proliferation of murine macrophages without significant cytotoxicity at concentrations up to 100 μM. In addition, the POH reduced the LPS-induced ROS and the expression of arginase-1 in M2-polarized macrophages.


Subject(s)
Animals , Mice , Fusobacterium nucleatum/drug effects , Reactive Oxygen Species/analysis , Porphyromonas/drug effects , Monoterpenes/pharmacology , Macrophages/drug effects , Anti-Bacterial Agents/pharmacology , Arginase/analysis , Time Factors , Biological Products/pharmacology , Microbial Sensitivity Tests , Gene Expression , Lipopolysaccharides/pharmacology , Reproducibility of Results , Tumor Necrosis Factor-alpha/analysis , Fusobacterium nucleatum/growth & development , Reactive Oxygen Species/metabolism , Porphyromonas/growth & development , Cell Proliferation/drug effects , Real-Time Polymerase Chain Reaction , Flow Cytometry , RAW 264.7 Cells , Macrophages/metabolism
8.
J Vet Sci ; 20(4): e33, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31364318

ABSTRACT

Porphyromonas species are closely associated with companion animal periodontitis which is one of the most common diseases in dogs and cats and leads to serious systemic diseases if left untreated. In this study, we evaluated the antimicrobial effects and mode of action of sodium tripolyphosphate (polyP3, Na5P3O10), a food additive with proven safety, using three pathogenic Porphyromonas species. The minimum inhibitory concentrations (MICs) of polyP3 against Porphyromonas gulae, Porphyromonas cansulci, and Porphyromonas cangingivalis were between 500 and 750 mg/L. PolyP3 significantly decreased viable planktonic cells as well as bacterial biofilm formation, even at sub-MIC concentrations. PolyP3 caused bacterial membrane disruption and this effect was most prominent in P. cangingivalis, which was demonstrated by measuring the amount of nucleotide leakage from the cells. To further investigate the mode of action of polyP3, high-throughput whole-transcriptome sequencing was performed using P. gulae. Approximately 30% of the total genes of P. gulae were differentially expressed by polyP3 (> 4-fold, adjusted p value < 0.01). PolyP3 influenced the expression of the P. gulae genes related to the biosynthesis of thiamine, ubiquinone, and peptidoglycan. Collectively, polyP3 has excellent antibacterial effects against pathogenic Porphyromonas species and can be a promising agent to control oral pathogenic bacteria in companion animals.


Subject(s)
Anti-Bacterial Agents/pharmacology , Polyphosphates/pharmacology , Porphyromonas/drug effects , Animals , Cat Diseases/microbiology , Cats , Dog Diseases/microbiology , Dogs , Periodontitis/microbiology , Periodontitis/veterinary , Species Specificity
9.
Article in English | MEDLINE | ID: mdl-29439961

ABSTRACT

Omadacycline (OMC), a broad-spectrum aminomethylcycline, has shown clinical efficacy in anaerobic acute bacterial skin and skin structure infections (ABSSSI) and in animal models of intra-abdominal anaerobic infections. Here, the in vitro activity of OMC against clinically relevant anaerobes was similar to that of tigecycline, with MIC90 values of 1 to 8 µg/ml against Bacteroides spp., 0.5 µg/ml against Clostridium difficile, Prevotella spp., and Porphyromonas asaccharolytica, 1 µg/ml against Peptostreptococcus spp., and 16 µg/ml against Clostridium perfringens.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria, Anaerobic/drug effects , Tetracyclines/pharmacology , Clostridioides difficile/drug effects , Microbial Sensitivity Tests , Peptostreptococcus/drug effects , Porphyromonas/drug effects , Prevotella/drug effects , Tigecycline/pharmacology
10.
Vet Microbiol ; 214: 132-139, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29408025

ABSTRACT

Broad-spectrum antibiotics such as ceftiofur and ampicillin are recommended for the treatment of metritis in dairy cows. Nonetheless, little is known about the impacts of antibiotics on the uterine microbiota. Here, we evaluated the shift in uterine microbiota after treating metritic cows with ceftiofur or ampicillin, and also gained insight into the uterine microbiota associated with cure of metritis. Uterine swabs from ceftiofur-treated, ampicillin-treated, and untreated metritic Holstein cows were collected on the day of metritis diagnosis (D1) and on D6 and then used for genomic DNA extraction and sequencing of the V4 hypervariable region of the 16S rRNA gene on the Illumina MiSeq platform. The uterine microbiota consolidated over time by decreasing species richness and increasing evenness; therefore, becoming more homogeneous. The uterine microbial community showed distinct clustering patterns on D6 according to antibiotic treatment, which could be attributed to more dynamic changes in the microbial structure from D1 to D6 in ceftiofur-treated cows. Ceftiofur led to significant changes at the community level, phylum level, and genus level, whereas the changes in ampicillin and untreated cows, although following the same pattern, were mostly non-significant. Bacteroidetes was significantly increased in ceftiofur-treated cows but was not changed after ampicillin and no treatment. Different responses to antibiotics were observed in Porphyromonas, which increased in relative abundance with ceftiofur and decreased with ampicillin. Regardless of treatment group, failure to cure metritis was associated with a decrease in diversity of uterine microbiota and an increase in the relative abundance of Bacteroides, Porphyromonas, and Fusobacterium.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Bacteria/isolation & purification , Endometritis/veterinary , Microbiota/drug effects , Uterus/microbiology , Ampicillin/administration & dosage , Ampicillin/therapeutic use , Animals , Anti-Bacterial Agents/administration & dosage , Bacteria/classification , Bacteria/drug effects , Bacteroidetes/drug effects , Bacteroidetes/isolation & purification , Cattle , Cephalosporins/administration & dosage , Cephalosporins/therapeutic use , Endometritis/drug therapy , Endometritis/microbiology , Female , Porphyromonas/drug effects , Porphyromonas/isolation & purification , Uterus/drug effects
11.
Article in English | MEDLINE | ID: mdl-28993327

ABSTRACT

Pexiganan, a cationic peptide, exhibited a broad range of anti-anaerobic antimicrobial activity. The MIC90s of studied isolates were as follows: Bacteroides fragilis, 16 µg/ml; other B. fragilis group spp., 4 µg/ml; Prevotella and Fusobacterium spp., 32 µg/ml; Porphyromonas spp., 64 µg/ml; Propionibacterium acnes, 4 µg/ml; Eggerthella lenta and Peptostreptococcus anaerobius, 32 µg/ml; other Gram-positive rods and cocci, 4 µg/ml; Clostridium perfringens, 128 µg/ml; and other clostridia, 256 µg/ml. Pexiganan cream shows potential as adjunctive therapy for skin and skin structure infections (SSSIs) involving anaerobes.


Subject(s)
Anaerobiosis/physiology , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Skin Diseases, Infectious/microbiology , Skin/microbiology , Actinobacteria/drug effects , Actinobacteria/growth & development , Actinobacteria/isolation & purification , Amino Acid Sequence , Anti-Bacterial Agents/chemical synthesis , Antimicrobial Cationic Peptides/chemical synthesis , Bacteroides fragilis/drug effects , Bacteroides fragilis/growth & development , Bacteroides fragilis/isolation & purification , Canada , Clostridium perfringens/drug effects , Clostridium perfringens/growth & development , Clostridium perfringens/isolation & purification , Firmicutes/drug effects , Firmicutes/growth & development , Firmicutes/isolation & purification , Fusobacterium/drug effects , Fusobacterium/growth & development , Fusobacterium/isolation & purification , Humans , Microbial Sensitivity Tests , Peptostreptococcus/drug effects , Peptostreptococcus/growth & development , Peptostreptococcus/isolation & purification , Porphyromonas/drug effects , Porphyromonas/growth & development , Porphyromonas/isolation & purification , Prevotella/drug effects , Prevotella/growth & development , Prevotella/isolation & purification , Propionibacterium acnes/drug effects , Propionibacterium acnes/growth & development , Propionibacterium acnes/isolation & purification , Skin/pathology , Skin Diseases, Infectious/pathology , Sweden , United States
12.
Future Med Chem ; 9(13): 1557-1574, 2017 09.
Article in English | MEDLINE | ID: mdl-28792235

ABSTRACT

AIM: Resistances to antibiotics employed for treatment of infectious diseases have increased to alarming numbers making it more and more difficult to treat diseases caused by microorganisms resistant to common antibiotics. Consequently, novel methods for successful inactivation of pathogens are required. In this instance, one alternative could be application of light for treatment of topical infections. Antimicrobial properties of UV light are well documented, but due to its DNA-damaging properties use for medical purposes is limited. In contrast, irradiation with visible light may be more promising. METHODS: Literature was systematically screened for research concerning inactivation of main oral bacterial species by means of visible light. RESULTS: Inactivation of bacterial species, especially pigmented ones, in planktonic state showed promising results. There is a lack of research examining the situation when organized as biofilms. CONCLUSION: More research concerning situation in a biofilm state is required.


Subject(s)
Anti-Infective Agents/pharmacology , Bacteria/drug effects , Light , Aggregatibacter/drug effects , Aggregatibacter/radiation effects , Anti-Infective Agents/chemistry , Bacteria/radiation effects , Escherichia coli/drug effects , Escherichia coli/radiation effects , Fusobacterium/drug effects , Fusobacterium/radiation effects , Humans , Mouth/microbiology , Porphyromonas/drug effects , Porphyromonas/radiation effects , Prevotella/drug effects , Prevotella/radiation effects , Staphylococcus/drug effects , Staphylococcus/radiation effects , Streptococcus/drug effects , Streptococcus/radiation effects
13.
BMC Complement Altern Med ; 17(1): 399, 2017 Aug 10.
Article in English | MEDLINE | ID: mdl-28797303

ABSTRACT

BACKGROUND: The common usage of chewing sticks prepared from Neem tree (Azadirachta indica) in India suggests its potential efficacy in periodontal diseases. The objective of this study is to explore the antibacterial effects of Neem leaf extract on the periodontophatic bacteria Porphyromonas gingivalis and Fusobacterium nucleatum, and its antioxidant capacities alone and in combination with bacteria and polycationic peptides that may be at the site of inflammation. METHODS: Neem leaf extract was prepared by ethanol extraction. The growth kinetics of P. gingivalis and F. nucleatum under anaerobic conditions in the presence of Neem leaf extract were measured. Broth microdilution test was used to determine the Minimal Inhibitory Concentration (MIC) of Neem leaf extract against each bacterial strain. The effect of Neem leaf extract on the coaggregation of the bacteria was assessed by a visual semi-quantitative assay. The antioxidant capacities of Neem leaf extract alone and in combination with bacteria, with the addition of red blood cells or the polycationic peptides chlorhexidine and lisozyme, were determined using a chemiluminescence assay. RESULTS: Neem leaf extract showed prominent dose-dependent antibacterial activity against P. gingivalis, however, had no effect on the growth of F. nucleatum nor on the coaggregation of the two bacteria. Yet, it showed intense antioxidant activity, which was amplified following adherence to bacteria and with the addition of red blood cells or the polycationic peptides. CONCLUSIONS: Neem leaf extract, containing polyphenols that adhere to oral surfaces, have the potential to provide long-lasting antibacterial as well as synergic antioxidant activities when in complex with bacteria, red blood cells and lisozyme. Thus, it might be especially effective in periodontal diseases.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antioxidants/pharmacology , Azadirachta/chemistry , Erythrocytes , Muramidase/metabolism , Periodontal Diseases/microbiology , Plant Extracts/pharmacology , Anti-Infective Agents, Local , Chlorhexidine , Fusobacterium/drug effects , Fusobacterium nucleatum/drug effects , Fusobacterium nucleatum/growth & development , Humans , India , Medicine, Traditional , Microbial Sensitivity Tests , Peptides , Periodontal Diseases/drug therapy , Periodontal Diseases/metabolism , Phytotherapy , Plant Leaves , Polyamines , Polyelectrolytes , Polyphenols/pharmacology , Porphyromonas/drug effects , Porphyromonas gingivalis/drug effects , Porphyromonas gingivalis/growth & development
14.
Anaerobe ; 47: 157-164, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28526497

ABSTRACT

Biofilms composed of anaerobic bacteria can result in persistent infections and chronic inflammation. Host immune cells have difficulties clearing biofilm-related infections and this can result in tissue damage. Neutrophils are a vital component of the innate immune system and help clear biofilms. The comparative neutrophilic response to biofilms versus planktonic bacteria remains incompletely understood, particularly in the context of mixed infections. The objective of this study was to generate mixed species anaerobic bacterial biofilms composed of two opportunistic pathogens, Fusobacterium necrophorum and Porphyromonas levii, and evaluate neutrophil responses to extracellular fractions from both biofilms and planktonic cell co-cultures of the same bacteria. Purified bovine neutrophils exposed to culture supernatants from mixed species planktonic bacteria showed elevated oxidative activity compared to neutrophils exposed to biofilms composed of the same bacteria. Bacterial lipopolysaccharide plays a significant role in the stimulation of neutrophils; biofilms produced substantially more lipopolysaccharide than planktonic bacteria under these experimental conditions. Removal of lipopolysaccharide significantly reduced neutrophil oxidative response to culture supernatants of planktonic bacteria. Oxidative responses to LPS-removed biofilm supernatants and LPS-removed planktonic cell supernatants were similar. The limited neutrophil response to biofilm bacteria observed in this study supports the reduced ability of the innate immune system to eradicate biofilm-associated infections. Lipopolysaccharide is likely important in neutrophil response; however, the presence of other extracellular, immune modifying molecules in the bacterial media also appears to be important in altering neutrophil function.


Subject(s)
Biofilms/growth & development , Fusobacterium necrophorum/immunology , Fusobacterium necrophorum/physiology , Neutrophils/immunology , Polysaccharides, Bacterial/metabolism , Porphyromonas/immunology , Porphyromonas/physiology , Animals , Cattle , Fusobacterium necrophorum/drug effects , Host-Pathogen Interactions , Neutrophils/drug effects , Oxidants/metabolism , Porphyromonas/drug effects
15.
Sci Rep ; 7: 43344, 2017 02 27.
Article in English | MEDLINE | ID: mdl-28240240

ABSTRACT

The numerous species that make up the oral microbiome are now understood to play a key role in establishment and maintenance of oral health. The ability to taxonomically identify community members at the species level is important to elucidating its diversity and association to health and disease. We report the overall ecological effects of using a toothpaste containing enzymes and proteins compared to a control toothpaste on the plaque microbiome. The results reported here demonstrate that a toothpaste containing enzymes and proteins can augment natural salivary defences to promote an overall community shift resulting in an increase in bacteria associated with gum health and a concomitant decrease in those associated with periodontal disease. Statistical analysis shows significant increases in 12 taxa associated with gum health including Neisseria spp. and a significant decrease in 10 taxa associated with periodontal disease including Treponema spp. The results demonstrate that a toothpaste containing enzymes and proteins can significantly shift the ecology of the oral microbiome (at species level) resulting in a community with a stronger association to health.


Subject(s)
Bacteria/drug effects , Dental Plaque/microbiology , Enzymes/pharmacology , Gingiva/microbiology , Microbiota/genetics , Mouth/metabolism , Toothpastes/pharmacology , Adolescent , Adult , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacterial Typing Techniques , Bacteroides/drug effects , Bacteroides/genetics , Bacteroides/isolation & purification , DNA, Bacterial/genetics , Female , Fusobacteria/drug effects , Fusobacteria/genetics , Fusobacteria/isolation & purification , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Oral Health , Oral Hygiene/methods , Porphyromonas/drug effects , Porphyromonas/genetics , Porphyromonas/isolation & purification , Prevotella/drug effects , Prevotella/genetics , Prevotella/isolation & purification , Selenomonas/drug effects , Selenomonas/genetics , Selenomonas/isolation & purification , Streptococcus/drug effects , Streptococcus/genetics , Streptococcus/isolation & purification , Treponema/drug effects , Treponema/genetics , Treponema/isolation & purification
16.
Carbohydr Polym ; 156: 417-426, 2017 Jan 20.
Article in English | MEDLINE | ID: mdl-27842841

ABSTRACT

This study aimed to compare two nanofiber drug delivery systems that were prepared with an electrospun process and have the potential to serve as adjuvants for the treatment of periodontal disease. The first system was composed of polycaprolactone loaded with tetracycline (TCN) and the second was composed of polycaprolactone loaded with tetracycline/ß-cyclodextrin (TCN:BCD). An antimicrobial diffusion test was performed for each of these sets of nanofibers with the microorganisms, Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis, both of which contribute to periodontal disease. In vitro release profiles were also obtained, and the nanofibers were characterized by thermal analysis, x-ray powder diffraction, infrared absorption spectroscopy, and scanning electron microscopy. Profiles of the TCN and TCN:BCD nanofibers showed that drug release occurred for up to 14days. However, the TCN:BCD nanofibers appeared to better protect and enhance the biological absorption of TCN due to the formation of a TCN:BCD inclusion complex.


Subject(s)
Aggregatibacter/drug effects , Nanofibers/chemistry , Porphyromonas/drug effects , Tetracycline/chemistry , Tetracycline/pharmacology , beta-Cyclodextrins/chemistry , beta-Cyclodextrins/pharmacology , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Microbial Sensitivity Tests
17.
Anaerobe ; 42: 37-39, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27510568

ABSTRACT

We report a first human case of Porphyromonas pogonae causing soft tissue infection in a patient with open fracture. Strong ß-hemolytic, aerotolerant, and non-pigmented gram-negative coccobacilli which matched Porphyromonas pogonae by PCR for 16S rRNA genes were identified from the pus specimen. The clinical course of the patient improved with repeated surgical drainage and tigecycline administration.


Subject(s)
Bacteroidaceae Infections/diagnosis , DNA, Bacterial/genetics , Fractures, Bone/diagnosis , Porphyromonas/isolation & purification , RNA, Ribosomal, 16S/genetics , Soft Tissue Infections/diagnosis , Adult , Anti-Bacterial Agents/therapeutic use , Bacteroidaceae Infections/drug therapy , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/surgery , Ciprofloxacin/therapeutic use , Fractures, Bone/drug therapy , Fractures, Bone/microbiology , Fractures, Bone/surgery , Humans , Male , Minocycline/analogs & derivatives , Minocycline/therapeutic use , Porphyromonas/classification , Porphyromonas/drug effects , Porphyromonas/genetics , Rifampin/therapeutic use , Sequence Analysis, DNA , Soft Tissue Infections/drug therapy , Soft Tissue Infections/microbiology , Soft Tissue Infections/surgery , Suppuration/microbiology , Tigecycline
18.
Bull Tokyo Dent Coll ; 57(3): 133-42, 2016.
Article in English | MEDLINE | ID: mdl-27665691

ABSTRACT

Periapical periodontitis usually results from microbial infection, with these microorganisms occasionally migrating to the root canal, which can lead to further, potentially life-threatening, complications. Here, the susceptibility of 27 bacterial strains to various antimicrobial agents was evaluated. These strains comprised 13 species; 16 of the strains were clinical isolates from periapical lesions. Each strain was inoculated onto blood agar plates containing one of the antimicrobial agents. The plates were incubated anaerobically at 37°C for 96 hr and the minimal inhibitory concentrations (MICs) determined. Ten strains required an MIC of 32 µg/ml or greater for amoxicillin, 6 for cefmetazole, and 5 for cefcapene among ß-lactam antibiotics; 8 strains required an MIC of 32 µg/ml or greater for clindamycin, 4 for azithromycin, and 11 for clarithromycin among macrolide antibiotics; 3 strains required an MIC of 32 µg/ml or greater for ciprofloxacin and 2 for ofloxacin among fluoroquinolones. The effect of cefcapene on 5 strains was evaluated after biofilm formation to investigate the relationship between biofilm formation and susceptibility. All strains showed a decrease in susceptibility after biofilm formation. The results revealed that several antimicrobial agents commonly used in a clinical setting, including amoxicillin, cefmetazole, and clindamycin, are potentially effective in the treatment of orofacial odontogenic infections. The development of resistant strains, however, means that this can no longer be guaranteed. In addition, azithromycin, ciprofloxacin, and ofloxacin were more effective than the 3 ß-lactam antibiotics tested. These results suggest that sensitivity testing is needed if odontogenic infections are to be treated safely and effectively.


Subject(s)
Drug Resistance, Bacterial , Periapical Periodontitis/microbiology , Actinomyces/drug effects , Amoxicillin/pharmacology , Anti-Bacterial Agents/pharmacology , Azithromycin/pharmacology , Biofilms/drug effects , Campylobacter/drug effects , Cefmetazole/pharmacology , Cephalosporins/pharmacology , Ciprofloxacin/pharmacology , Clarithromycin/pharmacology , Clindamycin/pharmacology , Fusobacteria/drug effects , Haemophilus/drug effects , Humans , Klebsiella/drug effects , Microbial Sensitivity Tests , Ofloxacin/pharmacology , Porphyromonas/drug effects , Propionibacterium/drug effects , Staphylococcus hominis/drug effects , Veillonella/drug effects
19.
Fitoterapia ; 83(6): 996-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22698715

ABSTRACT

The compounds 2',6'-dihydroxy-4'-geranyloxyacetophenone (1) and 2',6'-dihydroxy-4'-farnesyloxy-acetophenone (2) are oxyprenylated secondary metabolites extracted from plants belonging to the Rutaceae family. In this study, 1 and 2 were synthesized and tested for their antimicrobial activity toward major oral pathogens. Compounds 1 and 2 were synthesized by selective prenylation of 2,4,6-trihydroxyacetophenone at the 4' position with geranyl and farnesyl bromide, respectively. Compound 1 showed stronger antimicrobial activity than 2 against major oral pathogens, including Gram positive bacteria (Streptococcus mutans, Streptococcus sobrinus), Gram negative bacteria (Prevotella intermedia, Porphyromonas gingivalis) and Candida albicans. Evidences were obtained that the mode of action of 1 and 2 may be related to their iron-chelating property. This study suggests that 1 and 2 may represent potential natural molecules for the prevention/treatment of common oral infections, including dental caries, periodontal disease, and candidiasis.


Subject(s)
Acetophenones/pharmacology , Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Mouth/microbiology , Plant Extracts/pharmacology , Rutaceae/chemistry , Acetophenones/chemical synthesis , Acetophenones/chemistry , Acetophenones/therapeutic use , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/therapeutic use , Antifungal Agents/chemical synthesis , Antifungal Agents/therapeutic use , Bacteria/drug effects , Candida albicans/drug effects , Candidiasis/drug therapy , Chelating Agents/chemical synthesis , Chelating Agents/pharmacology , Chelating Agents/therapeutic use , Dental Caries/drug therapy , Iron/metabolism , Microbial Sensitivity Tests , Periodontal Diseases/drug therapy , Phytotherapy , Plant Extracts/chemistry , Plant Extracts/therapeutic use , Porphyromonas/drug effects , Prenylation , Prevotella/drug effects , Streptococcus/drug effects
20.
Anaerobe ; 18(4): 381-5, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22609780

ABSTRACT

The occurrence of Porphyromonas gulae, Porphyromonas macacae, Fusobacterium nucleatum and Fusobacterium canifelinum in subgingival plaque from dogs with and without periodontitis as well as their antimicrobial susceptibility were evaluated. From 50 dogs with periodontitis were identified 38 P. gulae, 8 P. macacae, 26 F. nucleatum and 15 F. canifelinum, and from 50 dogs without periodontitis were identified 15 P. gulae, 12 F. nucleatum and 11 F. canifelinum. All strains were susceptible to most of the antibiotics tested, however, different resistance rates to clarithromycin, erythromycin and metronidazole among strains were observed. The role of P. gulae, P. macacae, F. nucleatum and F. canifelinum in periodontal disease of household pets needs to be defined to a better prevention and treatment of the canine periodontitis.


Subject(s)
Dog Diseases/microbiology , Dogs/microbiology , Fusobacterium/drug effects , Fusobacterium/isolation & purification , Periodontitis/microbiology , Porphyromonas/drug effects , Porphyromonas/isolation & purification , Animals , Anti-Bacterial Agents/pharmacology , Bacteroidaceae Infections/microbiology , Clarithromycin/pharmacology , Dental Plaque/microbiology , Drug Resistance, Bacterial , Erythromycin/pharmacology , Female , Fusobacterium/metabolism , Fusobacterium Infections/microbiology , Hemagglutination Inhibition Tests , Humans , Male , Metronidazole/pharmacology , Microbial Sensitivity Tests , Porphyromonas/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...