Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 155
1.
J Med Case Rep ; 18(1): 213, 2024 Apr 30.
Article En | MEDLINE | ID: mdl-38685076

BACKGROUND: Prevotella heparinolytica is a Gram-negative bacterium that is commonly found in the oral, intestinal, and urinary tracts. It has been extensively studied in lower respiratory tract infections in horses, which has heparinolytic activity and can secrete heparinase and further induces virulence factors in cells and causes disease. However, no such cases have been reported in humans. CASE PRESENTATION: A 58-year-old male patient from China presented to the respiratory clinic in Suzhou with a productive cough producing white sputum for 20 days and fever for 3 days. Prior to this visit, a chest computed tomography scan was conducted, which revealed multiple patchy nodular opacities in both lungs. On admission, the patient presented with a temperature of 38.1 °C and a pulse rate of 110 beats per minute. Despite routine anti-infective treatment with moxifloxacin, his temperature fluctuated and the treatment was ineffective. The patient was diagnosed with Prevotella heparinolytica infection through metagenomic next-generation sequencing. Therefore, the antibiotics were switched to piperacillin-tazobactam in combination with ornidazole, which alleviated his symptoms; 1 week after discharge, the patient returned to the clinic for a follow-up chest computed tomography, and the opacities on the lungs continued to be absorbed. CONCLUSION: Prevotella heparinolytica is an opportunistic pathogen. However, it has not been reported in human pneumonia. In refractory pneumonia, measures such as metagenomic next-generation sequencing can be used to identify pathogens and help guide antibiotic selection and early support.


Anti-Bacterial Agents , Prevotella , Tomography, X-Ray Computed , Humans , Male , Middle Aged , Prevotella/isolation & purification , Anti-Bacterial Agents/therapeutic use , Bacteroidaceae Infections/drug therapy , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/diagnosis , Pneumonia, Bacterial/drug therapy , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/diagnosis , Piperacillin, Tazobactam Drug Combination/therapeutic use
2.
J Virol ; 95(18): e0047921, 2021 08 25.
Article En | MEDLINE | ID: mdl-34232744

HIV-1 elite controllers (EC) are a rare group among HIV-1-infected individuals who can naturally control viral replication for a prolonged period. Due to their heterogeneous nature, no universal mechanism could be attributed to the EC status; instead, several host and viral factors have been discussed as playing a role. In this study, we investigated the fecal metabolome and microbiome in a Swedish cohort of EC (n = 14), treatment-naive viremic progressors (VP; n = 16), and HIV-negative individuals (HC; n = 12). Fecal untargeted metabolomics was performed by four ultra-high-performance liquid chromatography tandem mass spectrometry (UHPLC-MS/MS). Molecular docking and biochemical microscale thermophoresis (MST) were used to describe the peptide-metabolite interactions. Single-cycle infectivity assays were performed in TZM-Bl cell lines using CCR5- and CXCR4-tropic HIV-1 strains. The microbiome analysis was performed using 16S rRNA sequencing. Th effects of metabolites on bacterial species viability were determined using several clinical isolates. We observed an enrichment of dipeptides in EC compared to VP and HC (adjusted P < 0.05). In silico analysis by molecular docking, in vitro biochemical assays, and ex vivo infection assays identified anti-HIV-1 properties for two dipeptides (WG and VQ) that could bind to the HIV-1 gp120, of which WG was more potent. The microbiome analysis identified enrichment of the genus Prevotella in EC, and these dipeptides supported bacterial growth of the genus Prevotella in vitro. The enrichments of the dipeptides and higher abundance of Prevotella have a distinct mechanism of elite control status in HIV-1 infection that influences host metabolism. IMPORTANCE HIV-1 elite controllers (EC) are a rare group among HIV-1-infected individuals who can naturally control viral replication for a prolonged period. Due to their heterogeneous nature, no universal mechanism could be attributed to the EC status; instead, several host and viral factors have been discussed as playing a role. In this study, we investigated the fecal metabolome and microbiome in a Swedish cohort of EC, treatment-naive viremic progressors (VP), and HIV-negative individuals (HC). We observed an enrichment of dipeptides in EC compared to the other two study groups. In silico and in vitro analyses identified anti-HIV-1 properties for two dipeptides that could bind to the HIV-1 gp120 and act as an HIV-1 antagonist. Furthermore, these dipeptides supported bacterial growth of the genus Prevotella in vitro that was enriched in EC, which influences host metabolism. Thus, increased levels of both dipeptides and Prevotella could provide beneficial effects for EC.


Bacteroidaceae Infections/microbiology , Dipeptides/pharmacology , Feces/microbiology , HIV Infections/prevention & control , HIV-1/physiology , Metabolome , Prevotella/pathogenicity , Adult , Bacteroidaceae Infections/drug therapy , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Case-Control Studies , Cohort Studies , Feces/chemistry , Female , Gene Expression Profiling , HIV Infections/virology , HIV-1/drug effects , Humans , Male , Middle Aged , Molecular Docking Simulation , Phenotype , Virus Replication
3.
BMC Anesthesiol ; 21(1): 102, 2021 04 05.
Article En | MEDLINE | ID: mdl-33820534

BACKGROUND: Laryngeal mask airways (LMA) are commonly used for airway management. Complications with this device are rare. However, when they do occur, there is a high risk for respiratory problems, necessitating early diagnosis and treatment. We present the first case of a life-threatening abscess spreading in the visceral space caused by a penicillin and metronidazole resistant Prevotella Denticola after the use of an LMA. CASE PRESENTATION: A female patient was admitted to our day care centre for bunion surgery. A single use LMA size 3 (Solus®, intersurgical, Wokingham, Berkshire, United Kingdom) was successfully inserted. After surgery, the patient complained of a sore throat and amoxicillin was prescribed by the general practitioner. Three days after surgery the patient was admitted to the Intensive Care Unit (ICU) for obstructive breathing, due to an abscess in the visceral space. Retropharyngeal and certainly parapharyngeal abscesses in adults are already rare. This case however, is unique because it is the first case of abscess spreading into the visceral space after the use of an LMA. Amoxicillin/clavulanate and vancomycin were started. The abscess was incised 5 days later and microbiology showed 3 positive cultures of the anaerobe Prevotella denticola, resistant for penicillin and metronidazole, but sensitive for amoxicillin/clavulanate. The patient fully recovered. CONCLUSION: LMA's are easy to use and are established, safe tools to support ventilation of the airway. In this case, the authors hypothesise a small wound in the lateral pharyngeal wall probably created an opening into the visceral space causing infection with Prevotella denticola, supporting the idea that the pharyngeal mucosal space must be part of the visceral space. Additionally, early recognition and treatment of an LMA induced abscess is necessary to prevent evolution of complications leading to airway obstruction.


Abscess/microbiology , Bacteroidaceae Infections/drug therapy , Laryngeal Masks/adverse effects , Abscess/drug therapy , Amoxicillin-Potassium Clavulanate Combination/therapeutic use , Anti-Bacterial Agents/therapeutic use , Drug Resistance, Multiple, Bacterial , Female , Humans , Middle Aged , Prevotella , Vancomycin/therapeutic use
4.
Anaerobe ; 69: 102346, 2021 Jun.
Article En | MEDLINE | ID: mdl-33600958

This study aimed to evaluate the antimicrobial activity of a new quinolone, lascufloxacin, for the treatment of complicated pneumonia caused by Streptococcus pneumoniae and Prevotella intermedia using a neutropenic mice pneumonia mixed-infection model. In this study, one S. pneumoniae and four P. intermedia isolates were utilized. Antimicrobial efficacy was calculated for each isolate as the reduction of the bacterial count comparatively to the non-treated mice (log10 colony forming units (cfu)/mL) obtained in the lungs of the treated mice after 24 h. Consequently, the bacterial densities of S. pneumoniae (KY-9) and P. intermedia (335) in the lungs of control animals were 8.20 ± 0.19 log10 cfu/mL and 5.26 ± 1.50 log10 cfu/mL, respectively. At human-simulated doses, lascufloxacin and levofloxacin showed high antimicrobial activities against not only S. pneumoniae (lascufloxacin: 1.88 ± 0.43 log10 cfu/mL, p < 0.001; levofloxacin 4.30 ± 0.75 log10 cfu/mL, p < 0.001), but also P. intermedia (lascufloxacin: 1.54 ± 0.57 log10 cfu/mL, p < 0.001; levofloxacin: 2.79 ± 0.55 log10 cfu/mL, p = 0.0102). Additionally, levofloxacin demonstrated attenuated antimicrobial efficacies against S. pneumoniae in the mixed-infection model compared with that in the single infection model. In contrast, lascufloxacin showed enhanced antimicrobial activities against S. pneumoniae and P. intermedia in the mixed-infection model. In conclusion, lascufloxacin resulted in enhanced efficacies against S. pneumoniae and P. intermedia, in both the single and mixed-infection models used. These data support the clinical utility of lascufloxacin for use against S. pneumoniae and P. intermedia in the treatment of pneumonia.


Anti-Bacterial Agents/pharmacology , Bacteroidaceae Infections/drug therapy , Fluoroquinolones/pharmacology , Levofloxacin/pharmacology , Pneumococcal Infections/drug therapy , Pneumonia/drug therapy , Prevotella intermedia/drug effects , Streptococcus pneumoniae/drug effects , Animals , Coinfection/drug therapy , Disease Models, Animal , Humans , Mice , Microbial Sensitivity Tests , Pneumonia/physiopathology
5.
J Tissue Eng Regen Med ; 15(3): 232-243, 2021 03.
Article En | MEDLINE | ID: mdl-33434402

Infectious bone defects remain a significant challenge in orthopedics and dentistry. Calcium phosphate cement (CPC) have attracted significant interest in use as local drug delivery system, which with great potential to control release of antibiotics for the treatment of infectious bone defects. Within the current study, a novel antibacterial scaffold of chitosan-reinforced calcium phosphate cement delivering doxycycline hyclate (CPCC + DOX) was developed. Furthermore, the capacity of CPCC + DOX scaffolds for bone regeneration was enhanced by the human periodontal ligament stem cells (hPDLSCs) encapsulated in alginate beads. CPCC + DOX scaffolds were fabricated to contain different concentrations of DOX. Flexural strength of CPCC + DOX ranged from 5.56 ± 0.70 to 6.2 ± 0.72 MPa, which exceeded the reported strength of cancellous bone. Scaffolds exhibited continual DOX release, reaching 80% at 21 days. Scaffold with 5 mg/ml DOX (CPCC + DOX5mg) had a strong antibacterial effect, with a 4-log colony forming unit reduction against S. aureus and P. gingivalis. The proliferation and osteogenic differentiation of hPDLSCs encapsulated in alginate hydrogel microbeads were investigated in culture with CPCC + DOX scaffolds. CPCC + DOX5mg had no negative effect on proliferation of hPDLSCs. Alkaline phosphatase activity, mineral synthesis, and osteogenic gene expressions for CPCC + DOX5mg group were much higher than control group. DOX did not compromise the osteogenic induction. In summary, the novel CPCC + DOX scaffold exhibited excellent mechanical properties and strong antibacterial activity, while supporting the proliferation and osteogenic differentiation of hPDLSCs. The CPCC + DOX + hPDLSCs construct is promising to enhance bone regeneration and combat bone infections in dental, craniofacial, and orthopedic applications.


Anti-Bacterial Agents , Bacteroidaceae Infections , Bone Cements , Bone Regeneration/drug effects , Microspheres , Osteogenesis , Periodontal Ligament , Porphyromonas gingivalis/growth & development , Staphylococcal Infections , Staphylococcus aureus/growth & development , Stem Cells , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacteroidaceae Infections/drug therapy , Bacteroidaceae Infections/microbiology , Bone Cements/chemistry , Bone Cements/pharmacology , Calcium Phosphates , Humans , Periodontal Ligament/metabolism , Periodontal Ligament/microbiology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/metabolism , Stem Cells/metabolism , Stem Cells/microbiology
6.
Phytomedicine ; 81: 153420, 2021 Jan.
Article En | MEDLINE | ID: mdl-33278781

BACKGROUND: Periodontitis is a common oral disease characterized as inflammation on gingival tissue and alveolar bone resorption. Spirulina maxima has been reported to have anti-oxidative and anti-inflammatory effects on gastric ulcers. However, its effects on gingival inflammation and alveolar bone resorption of periodontitis have not been studied. PURPOSE: This study was designed to investigate the effects of S. maxima on the P. gingivalis-induced periodontitis and to elucidate its mechanism. METHODS: The phycocyanin contents in S. maxima were identified by high-performance liquid chromatography. 8-week old SD rats were induced periodontitis by inoculation with P. gingivalis for 14 days. The rats were then orally treated with S. maxima 100, 200, 400 mg/kg, or indomethacin (IND, positive control) 5 mg/kg for an additional 14 days. Inflammatory responses, expressions of collagenases in gingival tissue, osteoclast formation and activation, alveolar bone resorption, osteogenesis-related markers, and BMP2/Smad signaling in alveolar bone were measured. RESULTS: Pro-inflammatory cytokines such as TNF-α, IL-1ß, IL-6, and inflammatory transcription factor NF-κB were decreased in gingival tissue by S. maxima administration. Also, myeloperoxidase (MPO) activity and matrix metalloproteinase (MMPs) expression were decreased by S. maxima administration. Conversely, S. maxima increased IL-4, anti-inflammatory cytokine from Th2 cells. The osteoprotegerin (OPG) / receptor activator of NF-κB ligand (RANKL) expression ratio, which represents osteoclast-osteoblast balance, was increased in S. maxima-treated groups. The alveolar bone loss and the number of TRAP-positive osteoclast cells were also declined in S. maxima-treated groups while the osteoblasts count was increased. Besides, in S. maxima-treated groups, the osteogenesis-related factors were promoted and BMP-2/Smad pathway was up-regulated in a periodontitis condition. CONCLUSION: S. maxima reduces periodontitis induced by P. gingivalis through anti-inflammatory effect and resultant reduction in bone loss, suggesting that S. maxima might be a potential agent for treating periodontitis.


Alveolar Bone Loss/drug therapy , Bacteroidaceae Infections/complications , Periodontitis/drug therapy , Periodontitis/microbiology , Porphyromonas gingivalis , Spirulina/chemistry , Alveolar Bone Loss/metabolism , Alveolar Bone Loss/microbiology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bacteroidaceae Infections/drug therapy , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/drug effects , Male , Osteoblasts/drug effects , Osteoclasts/drug effects , Periodontitis/metabolism , Phycocyanin/analysis , Rats, Sprague-Dawley , Smad Proteins/metabolism
7.
Int J Immunopathol Pharmacol ; 34: 2058738420974893, 2020.
Article En | MEDLINE | ID: mdl-33259259

Periodontitis is an inflammatory disease caused by host immune response, resulting in a loss of periodontium and alveolar bone. Immune cells, such as T cells and macrophages, play a critical role in the periodontitis onset. Halofuginone, a natural quinazolinone alkaloid, has been shown to possess anti-fibrosis, anti-cancer, and immunomodulatory properties. However, the effect of halofuginone on periodontitis has never been reported. In this study, a ligature-induced mice model of periodontitis was applied to investigate the potential beneficial effect of halofuginone on periodontitis. We demonstrated that the administration of halofuginone significantly reduced the expression levels of pro-inflammatory cytokines (IL-1ß, IL-6, and TNF-α) in vivo, and markedly suppressed immune cell infiltration into the infected sites. Furthermore, we also observed that halofuginone treatment blocked the T-helper 17 (Th17) cell differentiation in vivo and in vitro. We demonstrated for the first time that halofuginone alleviated the onset of periodontitis through reducing immune responses.


Bacteroidaceae Infections/drug therapy , Chronic Periodontitis/drug therapy , Gingiva/drug effects , Immunologic Factors/pharmacology , Piperidines/pharmacology , Quinazolinones/pharmacology , Animals , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology , Cell Differentiation/drug effects , Cells, Cultured , Chronic Periodontitis/immunology , Chronic Periodontitis/metabolism , Chronic Periodontitis/microbiology , Cytokines/metabolism , Disease Models, Animal , Gingiva/immunology , Gingiva/metabolism , Gingiva/microbiology , Host-Pathogen Interactions , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Porphyromonas gingivalis/immunology , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism
8.
PLoS Pathog ; 16(10): e1008881, 2020 10.
Article En | MEDLINE | ID: mdl-33002094

Epidemiological studies reveal significant associations between periodontitis and oral cancer. However, knowledge about the contribution of periodontal pathogens to oral cancer and potential regulatory mechanisms involved is limited. Previously, we showed that nisin, a bacteriocin and commonly used food preservative, reduced oral cancer tumorigenesis and extended the life expectancy in tumor-bearing mice. In addition, nisin has antimicrobial effects on key periodontal pathogens. Thus, the purpose of this study was to test the hypothesis that key periodontal pathogens (Porphyromonas gingivalis, Treponema denticola, and Fusobacterium nucleatum) promote oral cancer via specific host-bacterial interactions, and that bacteriocin/nisin therapy may modulate these responses. All three periodontal pathogens enhanced oral squamous cell carcinoma (OSCC) cell migration, invasion, tumorsphere formation, and tumorigenesis in vivo, without significantly affecting cell proliferation or apoptosis. In contrast, oral commensal bacteria did not affect OSCC cell migration. Pathogen-enhanced OSCC cell migration was mediated via integrin alpha V and FAK activation, since stably blocking alpha V or FAK expression abrogated these effects. Nisin inhibited these pathogen-mediated processes. Further, Treponema denticola induced TLR2 and 4 and MyD88 expression. Stable suppression of MyD88 significantly inhibited Treponema denticola-induced FAK activation and abrogated pathogen-induced migration. Together, these data demonstrate that periodontal pathogens contribute to a highly aggressive cancer phenotype via crosstalk between TLR/MyD88 and integrin/FAK signaling. Nisin can modulate these pathogen-mediated effects, and thus has therapeutic potential as an antimicrobial and anti-tumorigenic agent.


Bacteroidaceae Infections/drug therapy , Carcinoma, Squamous Cell/drug therapy , Mouth Neoplasms/drug therapy , Periodontitis/drug therapy , Porphyromonas gingivalis/drug effects , Probiotics/pharmacology , Animals , Apoptosis , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/pathology , Cell Movement , Cell Proliferation , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Humans , Integrins/genetics , Integrins/metabolism , Mice , Mice, Nude , Mouth Neoplasms/metabolism , Mouth Neoplasms/microbiology , Mouth Neoplasms/pathology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Periodontitis/metabolism , Periodontitis/microbiology , Periodontitis/pathology , Porphyromonas gingivalis/pathogenicity , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Anaerobe ; 65: 102266, 2020 Oct.
Article En | MEDLINE | ID: mdl-32871243

Strong evidence points to Porphyromonas gingivalis, a Gram-negative anaerobic bacterium, as a keystone species in the development of the chronic form of periodontitis. The aim of the present study was to investigate the ability of highbush blueberry proanthocyanidins (PACs) to alleviate the P. gingivalis-induced deleterious effects on oral mucosal cells. We first showed that highbush blueberry PACs protect the integrity of the gingival keratinocyte barrier against P. gingivalis-mediated damage, as determined by measuring the transepithelial electrical resistance and paracellular flux of FITC-conjugated dextran. Moreover, the PACs prevented the translocation of P. gingivalis across the gingival keratinocyte barrier model. The proteinase activity of P. gingivalis was inhibited by the PACs suggesting that they may exert beneficial effects by reducing proteolytic degradation of the epithelial tight junctions. Regulation of gingival fibroblast inflammatory reactions may be one of the ways to prevent and control periodontal disease progression and severity. We showed that PACs significantly reduce IL-6 and IL-8 secretion by P. gingivalis-stimulated gingival fibroblasts. The present study showed the capacity of highbush blueberry PACs to protect the integrity of an in vitro model of gingival keratinocyte barrier against P. gingivalis, and to attenuate the secretion of pro-inflammatory cytokines by gingival fibroblasts infected with P. gingivalis. These results suggest beneficial effects of blueberry PACs thus supporting the need for future clinical trials on the potential of these bioactive molecules for periodontal disease prevention and/or treatment.


Bacteroidaceae Infections/microbiology , Blueberry Plants/chemistry , Periodontitis/microbiology , Porphyromonas gingivalis/drug effects , Proanthocyanidins/pharmacology , Bacteroidaceae Infections/drug therapy , Cells, Cultured , Cytokines/biosynthesis , Humans , Inflammation Mediators/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/microbiology , Mouth Mucosa/microbiology , Mouth Mucosa/pathology , Periodontitis/drug therapy , Periodontitis/pathology
10.
Sci Rep ; 10(1): 14745, 2020 09 08.
Article En | MEDLINE | ID: mdl-32901057

The control of inflammation and infection is crucial for periodontal wound healing and regeneration. M101, an oxygen carrier derived from Arenicola marina, was tested for its anti-inflammatory and anti-infectious potential based on its anti-oxidative and tissue oxygenation properties. In vitro, no cytotoxicity was observed in oral epithelial cells (EC) treated with M101. M101 (1 g/L) reduced significantly the gene expression of pro-inflammatory markers such as TNF-α, NF-κΒ and RANKL in P. gingivalis-LPS stimulated and P. gingivalis-infected EC. The proteome array revealed significant down-regulation of pro-inflammatory cytokines (IL-1ß and IL-8) and chemokine ligands (RANTES and IP-10), and upregulation of pro-healing mediators (PDGF-BB, TGF-ß1, IL-10, IL-2, IL-4, IL-11 and IL-15) and, extracellular and immune modulators (TIMP-2, M-CSF and ICAM-1). M101 significantly increased the gene expression of Resolvin-E1 receptor. Furthermore, M101 treatment reduced P. gingivalis biofilm growth over glass surface, observed with live/dead analysis and by decreased P. gingivalis 16 s rRNA expression (51.7%) (p < 0.05). In mice, M101 reduced the clinical abscess size (50.2%) in P. gingivalis-induced calvarial lesion concomitant with a decreased inflammatory score evaluated through histomorphometric analysis, thus, improving soft tissue and bone healing response. Therefore, M101 may be a novel therapeutic agent that could be beneficial in the management of P. gingivalis associated diseases.


Anti-Inflammatory Agents/pharmacology , Bacteroidaceae Infections/complications , Brain Abscess/drug therapy , Inflammation/drug therapy , Oxygen/pharmacology , Polychaeta/chemistry , Skull/drug effects , Animals , Bacteroidaceae Infections/drug therapy , Bacteroidaceae Infections/microbiology , Brain Abscess/microbiology , Brain Abscess/pathology , Gingiva/chemistry , Gingiva/microbiology , Humans , Inflammation/microbiology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Porphyromonas gingivalis/drug effects , Porphyromonas gingivalis/growth & development , Porphyromonas gingivalis/isolation & purification , Signal Transduction , Skull/microbiology , Skull/pathology
11.
BMC Complement Med Ther ; 20(1): 259, 2020 Aug 20.
Article En | MEDLINE | ID: mdl-32819379

BACKGROUND: The Sri Lankan traditional betel quid (TBQ) which had been extensively used in the country before its colonization is claimed to have antiperiodontopathic effects in the Sri Lankan folklore. However, there is no reported scientific evidence to support the claimed antiperiodontopathic effects mediated by this TBQ. The present study was carried out to investigate the protective effect of the Sri Lankan TBQ in the pathogenesis of periodontitis. METHODS: We investigate the ethyl acetate extract of the Sri Lankan TBQ for its antibacterial effects against the keystone periodontopathic bacterium, P. gingivalis and also its antioxidant potential, which is important to protect the periodontium from oxidative stress. Further, its safety was analyzed using the cytokinesis block micronucleus assay on human peripheral blood lymphocytes (PBLs). RESULTS: Ethyl acetate extract of this TBQ inhibited the growth of P. gingivalis with a minimum bactericidal concentration (MBC) of 125 µg/ml. It was found to be a rich source of polyphenols and displayed considerable DPPH and ABTS radical scavenging activities and a strong ferric reducing antioxidant power. This extract could protect the cultured human gingival fibroblasts from H2O2 induced oxidative stress. In addition, this TBQ extract was not genotoxic to human PBLs even at a concentration of 2.5 mg/ml. Moreover, it exhibited protective effects against bleomycin induced genotoxicity in PBLs. CONCLUSION: Ethyl acetate extract of the Sri Lankan TBQ is a source of natural antibacterial compounds against P. gingivalis. It is also a source of natural antioxidants which can protect human gingival fibroblasts from H2O2 induced oxidative stress. These properties of the TBQ may have contributed to its claimed antiperiodontopathic effects. Besides, it was found to be relatively non-toxic to human cells. Thus this TBQ extract has a huge potential to be developed as a novel adjunctive therapeutic lead against periodontitis.


Anti-Bacterial Agents/pharmacology , Bacteroidaceae Infections/drug therapy , Periodontitis/drug therapy , Piper betle , Plant Extracts/pharmacology , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Plant Leaves , Porphyromonas gingivalis , Sri Lanka
12.
Aging (Albany NY) ; 12(11): 10117-10128, 2020 06 09.
Article En | MEDLINE | ID: mdl-32516126

Amyloid-ß (Aß) accumulation is one of the main pathological hallmarks of Alzheimer's disease (AD). Porphyromonas gingivalis (P. gingivalis), the pathogen of chronic periodontitis, could cause Aß accumulation and was identified in the brain of AD patients. Salvianolic Acid B (SalB) has been proven to have the neuroprotective effect. Whether SalB could protect against P. gingivalis-induced cognitive impairment is still unknown. In this study, a P. gingivalis-infected mouse model was employed to study the neuroprotective role of SalB. The results showed that SalB (20 and 40 mg/kg) treatment for 4 weeks could shorten the escape latency and improve the percentage of spontaneous alternation in the P. gingivalis-infected mice. SalB inhibited the levels of reactive oxygen species and malondialdehyde, while increased the levels of antioxidative enzymes (superoxide dismutase and glutathione peroxidase). SalB decreased the levels of IL-1ß and IL-6, increased the mRNA levels of bdnf and ngf in the brain of P. gingivalis-infected mice. In addition, SalB obviously decreased the level of Aß. SalB elevated the protein expression of ADAM10, while downregulated BACE1 and PS1. SalB increased the protein expression of LRP1, while decreased RAGE. In conclusion, SalB could improve cognitive impairment by inhibiting neuroinflammation and decreasing Aß level in P. gingivalis-infected mice.


Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Bacteroidaceae Infections/complications , Benzofurans/pharmacology , Cognitive Dysfunction/drug therapy , Administration, Oral , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/immunology , Animals , Bacteroidaceae Infections/drug therapy , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/microbiology , Behavior Observation Techniques , Behavior, Animal/drug effects , Benzofurans/therapeutic use , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/immunology , Cognitive Dysfunction/pathology , Disease Models, Animal , Drugs, Chinese Herbal/therapeutic use , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Hippocampus/chemistry , Hippocampus/drug effects , Hippocampus/immunology , Hippocampus/pathology , Humans , Male , Malondialdehyde/analysis , Malondialdehyde/metabolism , Mice , Oxidative Stress/drug effects , Oxidative Stress/immunology , Porphyromonas gingivalis/immunology , Porphyromonas gingivalis/isolation & purification , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism
13.
PLoS Pathog ; 16(6): e1008559, 2020 06.
Article En | MEDLINE | ID: mdl-32497109

Antibiotics continue to be the standard-of-care for bacterial vaginosis (BV), although recurrence rates are high. Vaginal probiotics may improve durability of BV treatment, although few probiotics for vaginal health contain Lactobacillus spp. that commonly colonize the lower female genital tract. Characteristics of vaginal Lactobacillus strains from South African women were evaluated for their probiotic potential in vitro compared to strains from commercial vaginal products, including growth at varying pHs, ability to lower pH, produce D-/L-lactate and H2O2, influence growth of BV-associated Gardnerella vaginalis and Prevotella bivia, adherence to cervical cells and susceptibility to antibiotics. Fifty-seven Lactobacillus strains were purified from cervico-vaginal fluid, including L. crispatus, L. jensenii, L. gasseri, L. mucosae, and L. vaginalis. L crispatus strains grew better at pHs below 4.5 and lowered pH more effectively than other strains. Production of D-/L-lactate and H2O2 varied between Lactobacillus species and strains. Lactobacillus strains generally inhibited P. bivia more uniformly than G. vaginalis isolates. All vaginal Lactobacillus isolates were resistant to metronidazole while susceptibility to clindamycin varied. Furthermore, vaginal Lactobacillus strains tended to be broadly susceptible to penicillin, amoxicillin, rifampicin and rifabutin. Whole-genome-sequencing of five of the best-performing vaginal Lactobacillus strains confirmed their likely safety, due to antimicrobial resistance elements being largely absent, while putative intact prophages were present in the genomes of two of the five strains. Overall, vaginal Lactobacillus strains largely performed better in these in vitro assays than probiotic strains currently used in probiotics for vaginal health. Including the best-performing vaginal Lactobacillus isolates in a region-specific probiotic for vaginal health may result in improved BV treatment options.


Bacteroidaceae Infections/microbiology , Gardnerella vaginalis , Gram-Positive Bacterial Infections/microbiology , Lactobacillus , Prevotella , Vaginosis, Bacterial/microbiology , Adolescent , Adult , Bacteroidaceae Infections/drug therapy , Bacteroidaceae Infections/genetics , Bacteroidaceae Infections/metabolism , Clindamycin/pharmacology , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Female , Gram-Positive Bacterial Infections/drug therapy , Gram-Positive Bacterial Infections/genetics , Gram-Positive Bacterial Infections/metabolism , Humans , Hydrogen Peroxide/metabolism , Lactic Acid/metabolism , Lactobacillus/genetics , Lactobacillus/isolation & purification , Lactobacillus/metabolism , Metronidazole/pharmacology , South Africa , Species Specificity , Vaginosis, Bacterial/drug therapy , Vaginosis, Bacterial/genetics
14.
J Periodontol ; 91 Suppl 1: S45-S49, 2020 10.
Article En | MEDLINE | ID: mdl-32533852

Epidemiological studies have identified an association between periodontitis and Alzheimer disease (AD); however, the nature of this association has been unclear. Recent work suggests that brain colonization by the periodontal pathogen Porphyromonas gingivalis may link these two inflammatory and degenerative conditions. Evidence of P. gingivalis infiltration has been detected in autopsy specimens from the brains of people with AD and in cerebrospinal fluid of individuals diagnosed with AD. Gingipains, a class of P. gingivalis proteases, are found in association with neurons, tau tangles, and beta-amyloid in specimens from the brains of individuals with AD. The brains of mice orally infected with P. gingivalis show evidence of P. gingivalis infiltration, along with various neuropathological hallmarks of AD. Oral administration of gingipain inhibitors to mice with established brain infections decreases the abundance of P. gingivalis DNA in brain and mitigates the neurotoxic effects of P. gingivalis infection. Thus, gingipain inhibition could provide a potential approach to the treatment of both periodontitis and AD.


Alzheimer Disease , Bacteroidaceae Infections , Periodontitis , Adhesins, Bacterial , Alzheimer Disease/drug therapy , Animals , Bacteroidaceae Infections/drug therapy , Cysteine Endopeptidases , Humans , Mice , Porphyromonas gingivalis
15.
Sci Rep ; 10(1): 3113, 2020 02 20.
Article En | MEDLINE | ID: mdl-32080231

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.


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
16.
Pharmacol Res Perspect ; 8(1): e00562, 2020 02.
Article En | MEDLINE | ID: mdl-31999052

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.


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
17.
Nutrients ; 12(1)2020 Jan 20.
Article En | MEDLINE | ID: mdl-31968635

Periodontitis is a polymicrobial infectious disease that leads to inflammation of the gingiva, resulting in teeth loss by various causes such as inflammation-mediated bone resorption. Recently, many investigators have reported that the periodontitis resulting from persistent low-grade infection of Gram-negative bacteria such as Porphyromonas gingivalis (Pg) is associated with increased atherosclerosis, diabetes mellitus, and other systemic diseases through blood stream. On the other hand, carotenoids belong among phytochemicals that are responsible for different colors of the foods. It is important to examine whether carotenoids are effective to the inhibition of periodontal infection/inflammation cascades. This review summarizes the advanced state of knowledge about suppression of periodontal infection by several carotenoids. A series of findings suggest that carotenoids intake may provide novel strategy for periodontitis treatment, although further study will be needed.


Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Bacteroidaceae Infections/drug therapy , Carotenoids/therapeutic use , Periodontitis/drug therapy , Porphyromonas gingivalis/pathogenicity , Animals , Anti-Inflammatory Agents/adverse effects , Antioxidants/adverse effects , Bacteroidaceae Infections/microbiology , Carotenoids/adverse effects , Humans , Inflammation Mediators/metabolism , Oxidative Stress/drug effects , Periodontitis/microbiology , Reactive Oxygen Species/metabolism , Signal Transduction , Treatment Outcome
19.
Indian J Med Microbiol ; 37(1): 54-59, 2019.
Article En | MEDLINE | ID: mdl-31424011

Background: Porphyromonas gingivalis is a major periodontal pathogen. Saliva is the most easy, non-invasive microbiological sample for detection of periodontal pathogens. Aim and Objectives: A prospective study on 37 diabetic patients was grouped into well-controlled diabetes with/without periodontitis and uncontrolled diabetic with periodontitis. PCR and sequencing of P. gingivalis was performed in saliva samples. Materials and Methods: DNA was extracted from saliva using Triton X-100 and 16s rRNA gene (404 bp) was amplified by polymerase chain reaction. DNA sequencing was performed for two samples. Results: P. gingivalis was detected in 27.03% (n = 10), of which 30% (n = 9) were diabetic with periodontal disease and 14.3% (n = 1) were diabetic without periodontal disease. The percentage of poor oral hygiene was 50% and 20% in uncontrolled and controlled glycaemic patients, respectively. DNA sequencing of two samples showed 100% identity with the sequences in the GenBank database (Gen Bank accession no: KX640913-KX640914). Conclusion: Type 2 diabetes mellitus and periodontitis are interlinked. Early detection of P. gingivalis and appropriate treatment with doxycycline will also assist in controlling the glycaemic status.


Diabetes Complications/microbiology , Diabetes Mellitus, Type 2/epidemiology , Periodontitis/epidemiology , Porphyromonas gingivalis/genetics , Saliva/microbiology , Adult , Aged , Bacteroidaceae Infections/drug therapy , Bacteroidaceae Infections/transmission , Diabetes Mellitus, Type 2/pathology , Doxycycline/therapeutic use , Female , Glycated Hemoglobin/analysis , Glycemic Index/drug effects , Humans , India/epidemiology , Male , Middle Aged , Oral Hygiene/statistics & numerical data , Periodontitis/drug therapy , Periodontitis/microbiology , Polymerase Chain Reaction , Porphyromonas gingivalis/drug effects , Prospective Studies , RNA, Ribosomal, 16S/genetics
20.
Mol Pharm ; 16(9): 4017-4023, 2019 09 03.
Article En | MEDLINE | ID: mdl-31361146

The in vivo antibacterial activity of NO-releasing hyperbranched polymers was evaluated against Porphyromonas gingivalis, a key oral pathogen associated with periodontitis, using a murine subcutaneous chamber model. Escalating doses of NO-releasing polymers (1.5, 7.5, and 37.5 mg/kg) were administered into a P. gingivalis-infected chamber once a day for 3 days. Chamber fluids were collected on day 4, with microbiological evaluation indicating a dose-dependent bactericidal action. In particular, NO-releasing polymers at 37.5 mg/kg (1170 µg of NO/kg) achieved complete bacterial eradication (>6-log reduction in bacterial viability), demonstrating greater efficacy than amoxicillin (∼4-log reduction in bacterial viability), a commonly used antibiotic. Time-kill assays further revealed that largest dose (37.5 mg/kg; 1170 µg of NO/kg) resulted in ∼3-log killing of P. gingivalis after only a single dose. Based on these results, the potential clinical utility of NO-releasing hyperbranched polymers appears promising, particularly for oral health applications.


Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Bacteroidaceae Infections/drug therapy , Nitric Oxide/chemistry , Nitric Oxide/therapeutic use , Periodontitis/drug therapy , Polymers/chemistry , Porphyromonas gingivalis/drug effects , Amoxicillin/therapeutic use , Animals , Bacteroidaceae Infections/microbiology , Disease Models, Animal , Epoxy Compounds/chemistry , Male , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Microbial Viability/drug effects , Periodontitis/microbiology , Polyamines/chemistry , Treatment Outcome
...