RESUMO
Introduction. Peri-implantitis is a plaque-associated disease that leads to implant loss and arises from bacterial biofilms on the surface of the implant. Smoking is a risk factor for peri-implantitis and impedes treatment effectiveness. Additionally, aryl hydrocarbon receptor (AHR), IL-6, and IL-22 levels are related to peri-implantitis.Aim. We aimed to investigate the effects of nicotine on inflammatory response, bacterial growth and biofilm formation.Hypothesis/Gap Statement. We hypothesized that nicotine promoted pathogenic bacterial growth and biofilm formation, thereby aggravating inflammation.Methodology. The expression of AHR, IL-6 and IL-22 was measured in peri-implant sulci fluid using quantitative PCR and Western blot analyses. The cementum was incubated with bacterial suspension including Porphyromonas gingivalis, Streptococcus sanguinis and Fusobacterium nucleatum and treated with 100, 200, 250 and 300 µg ml-1 nicotine, and then, the absorbance and number of colony-forming units were detected. Biofilm formation was evaluated using the tissue culture plate method and safranin O staining. Carbohydrates and proteins were measured by the phenol-sulfuric acid method and the bicinchoninic acid method, respectively.Results. The results indicated that smoking increased the levels of AHR, IL-6 and IL-22. Functionally, nicotine promoted the growth of P. gingivalis, S. sanguinis and F. nucleatum. Additionally, it promoted the biofilm formation of these bacteria and increased the contents of carbohydrates and proteins.Conclusion. Nicotine promoted bacterial growth and biofilm build-up, suggesting that smoking may aggravate the progression of peri-implantitis.
Assuntos
Biofilmes , Nicotina , Peri-Implantite , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Nicotina/farmacologia , Humanos , Peri-Implantite/microbiologia , Fusobacterium nucleatum/efeitos dos fármacos , Fusobacterium nucleatum/crescimento & desenvolvimento , Fusobacterium nucleatum/fisiologia , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/crescimento & desenvolvimento , Masculino , Implantes Dentários/microbiologia , Feminino , Interleucina-6/metabolismo , Pessoa de Meia-Idade , Interleucinas/metabolismo , Streptococcus sanguis/efeitos dos fármacos , Streptococcus sanguis/crescimento & desenvolvimento , Bactérias/efeitos dos fármacos , Bactérias/classificação , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Bactérias/isolamento & purificação , Fumar/efeitos adversosRESUMO
BACKGROUND: Given the limited treatment options available for oral lichen planus (OLP), a study was undertaken to obtain preliminary information on the therapeutic efficacy of tinidazole mouth rinse in patients with OLP. METHODS: A prospective, open-label pilot study was conducted to assess the efficacy of thrice-daily tinidazole mouth rinse for one week in OLP patients (n = 27). Reticulation/erythema/ulceration (REU) scores and visual analog scale (VAS) scores were used to measure lesions at baseline and after one week of treatment. Mucosal samples were collected, and the abundance of Fusobacterium nucleatum was quantified using RT-PCR. Statistical analysis using t-test, Wilcoxon signed rank test and Pearson correlation test. RESULTS: After treatment, VAS scores significantly decreased in both reticular (P = 0.03) and erosive OLP patients (P = 0.003). However, REU scores significantly decreased only in erosive OLP patients (P = 0.002). The relative abundance of Fusobacterium nucleatum on the damaged mucosa surface significantly decreased in all OLP patients (P = 0.01). In erosive OLP patients, the triamcinolone group showed a significantly greater improvement in VAS scores compared to the tinidazole group (P = 0.01). However, there was no statistically significant correlation between the relative abundance of Fusobacterium nucleatum and REU scores in OLP patients (r = 0.0754, P = 0.61). CONCLUSION: Tinidazole mouth rinse showed potential in reducing disease severity in OLP patients and was well-tolerated, suggesting its viability as a local therapeutic option. However, randomized controlled studies are warranted to confirm these preliminary findings.
Assuntos
Fusobacterium nucleatum , Líquen Plano Bucal , Antissépticos Bucais , Tinidazol , Humanos , Projetos Piloto , Masculino , Feminino , Líquen Plano Bucal/tratamento farmacológico , Antissépticos Bucais/uso terapêutico , Tinidazol/uso terapêutico , Pessoa de Meia-Idade , Estudos Prospectivos , Fusobacterium nucleatum/efeitos dos fármacos , Idoso , Adulto , Resultado do TratamentoRESUMO
The dental implant market has experienced explosive growth, owing to the widespread acceptance of implants as the core of oral rehabilitation. Clinically, achieving simultaneous anti-infective effects and rapid osseointegration is a crucial but challenging task for implants. The demand for implants with long-term broad-spectrum antibacterial and immune-osteogenic properties is growing. Existing methods are limited by a lack of safety, efficiency, short-lasting anti-infective ability, and inadequate consideration of the immunomodulatory effects on osteogenesis. Herein, a ZnO/black TiO2-x heterojunction surface structure was designed as a near-infrared (NIR) light-responsive nanofilm immobilized on a titanium (Ti) implant surface. This nanofilm introduces abundant oxygen vacancies and heterojunctions, which enhance the photothermal and photoelectric abilities of Ti implants under NIR illumination by narrowing the band gap and improving interfacial charge transfer. The "photo-thermo-electric" implant exhibits excellent broad-spectrum antibacterial efficacy against three dental pathogenic bacteria (Porphyromonas gingivalis, Fusobacterium nucleatum, and Staphylococcus aureus, >99.4%) by destroying the bacterial membrane and increasing the production of intracellular reactive oxygen species. Additionally, the implant can effectively eliminate mature multispecies biofilms and kill bacteria inside the biofilms under NIR irradiation. Meanwhile, this implant can also induce the pro-regenerative transformation of macrophages and promote osteoblast proliferation and differentiation. Moreover, in vivo results confirmed the superior antibacterial and osteoimmunomodulatory properties of this dental implant. RNA sequencing revealed that the underlying osteogenic mechanisms involve activation of the Wnt/ß-catenin signaling pathway and bone development. Overall, this versatile "photo-thermo-electric" platform endows implants with anti-infection and bone integration performance simultaneously, which holds great potential for dental implants.
Assuntos
Antibacterianos , Implantes Dentários , Porphyromonas gingivalis , Staphylococcus aureus , Titânio , Implantes Dentários/microbiologia , Titânio/química , Titânio/farmacologia , Animais , Staphylococcus aureus/efeitos dos fármacos , Porphyromonas gingivalis/efeitos dos fármacos , Camundongos , Antibacterianos/farmacologia , Antibacterianos/química , Fusobacterium nucleatum/efeitos dos fármacos , Osseointegração/efeitos dos fármacos , Óxido de Zinco/química , Óxido de Zinco/farmacologia , Imunomodulação/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Propriedades de Superfície , Testes de Sensibilidade Microbiana , Osteogênese/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Células RAW 264.7 , Raios InfravermelhosRESUMO
OBJECTIVE: Epigallocatechin-3-gallate (EGCG), a catechin abundant in green tea, exhibits antibacterial activity. In this study, the antimicrobial effects of EGCG on periodontal disease-associated bacteria (Porphyromonas gingivalis, Prevotella intermedia, Prevotella nigrescens, Fusobacterium nucleatum, and Fusobacterium periodontium) were evaluated and compared with its effects on Streptococcus mutans, a caries-associated bacterium. RESULTS: Treatment with 2 mg/ml EGCG for 4 h killed all periodontal disease-associated bacteria, whereas it only reduced the viable count of S. mutans by about 40 %. Regarding growth, the periodontal disease-associated bacteria were more susceptible to EGCG than S. mutans, based on the growth inhibition ring test. As for metabolism, the 50 % inhibitory concentration (IC50) of EGCG for bacterial metabolic activity was lower for periodontal disease-associated bacteria (0.32-0.65 mg/ml) than for S. mutans (1.14 mg/ml). Furthermore, these IC50 values were negatively correlated with the growth inhibition ring (r = -0.73 to -0.86). EGCG induced bacterial aggregation at the following concentrations: P. gingivalis (>0.125 mg/ml), F. periodonticum (>0.5 mg/ml), F. nucleatum (>1 mg/ml), and P. nigrescens (>2 mg/ml). S. mutans aggregated at an EGCG concentration of > 1 mg/ml. CONCLUSION: EGCG may help to prevent periodontal disease by killing bacteria, inhibiting bacterial growth by suppressing bacterial metabolic activity, and removing bacteria through aggregation.
Assuntos
Catequina , Fusobacterium nucleatum , Doenças Periodontais , Porphyromonas gingivalis , Prevotella intermedia , Streptococcus mutans , Chá , Catequina/farmacologia , Catequina/análogos & derivados , Chá/química , Streptococcus mutans/efeitos dos fármacos , Doenças Periodontais/microbiologia , Doenças Periodontais/tratamento farmacológico , Porphyromonas gingivalis/efeitos dos fármacos , Fusobacterium nucleatum/efeitos dos fármacos , Prevotella intermedia/efeitos dos fármacos , Fusobacterium/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Antibacterianos/farmacologia , Prevotella nigrescens/efeitos dos fármacos , HumanosRESUMO
Nano-mupirocin is a PEGylated nano-liposomal formulation of the antibiotic mupirocin, undergoing evaluation for treating infectious diseases and intratumor bacteria. Intratumoral microbiota play an important role in the regulation of tumor progression and therapeutic efficacy. However, antibiotic use to target intratumoral bacteria should be performed in a way that will not affect the gut microbiota, found to enable the efficacy of cancer treatments. Nano-mupirocin may offer such a selective treatment. Herein, we demonstrate the ability of Nano-mupirocin to successfully target tumor-residing Fusobacterium nucleatum without an immediate effect on the gut microbiome. In-depth characterization of this novel formulation was performed, and the main findings include: (i). the pharmacokinetic analysis of mupirocin administered as Nano-mupirocin vs mupirocin lithium (free drug) demonstrated that most of the Nano-mupirocin in plasma is liposome associated; (ii). microbiome analysis of rat feces showed no significant short-term difference between Nano-mupirocin, mupirocin lithium and controls; (iii). Nano-mupirocin was active against intratumoral F. nucleatum, a tumor promoting bacteria that accumulates in tumors of the AT3 mice model of breast cancer. These data suggest the ability of Nano-mupirocin to target tumor residing and promoting bacteria.
Assuntos
Antibacterianos , Fusobacterium nucleatum , Microbioma Gastrointestinal , Animais , Microbioma Gastrointestinal/efeitos dos fármacos , Antibacterianos/farmacocinética , Antibacterianos/administração & dosagem , Fusobacterium nucleatum/efeitos dos fármacos , Feminino , Lipossomos , Nanopartículas/química , Linhagem Celular Tumoral , Camundongos , Ratos Sprague-Dawley , Ratos , Fezes/microbiologia , Humanos , Polietilenoglicóis/químicaRESUMO
The performance and long-term durability of dental implants hinge on the quality of bone integration and their resistance to bacteria. This research aims to introduce a surface modification strategy for zirconia implants utilizing femtosecond laser ablation techniques, exploring their impact on osteoblast cell behavior and bacterial performance, as well as the integral factors influencing the soft tissue quality surrounding dental implants. Ultrafast lasers were employed to craft nanoscale groove geometries on zirconia surfaces, with thorough analyses conducted using x-ray diffraction, scanning electron microscopy, atomic force microscopy, and water contact angle measurements. The study evaluated the response of human fetal osteoblastic cell lines to textured zirconia ceramics by assessing alkaline phosphatase activity, collagen I, and interleukin 1ßsecretion over a 7 day period. Additionally, the antibacterial behavior of the textured surfaces was investigated usingFusobacterium nucleatum, a common culprit in infections associated with dental implants. Ciprofloxacin (CIP), a widely used antibacterial antibiotic, was loaded onto zirconia ceramic surfaces. The results of this study unveiled a substantial reduction in bacterial adhesion on textured zirconia surfaces. The fine biocompatibility of these surfaces was confirmed through the MTT assay and observations of cell morphology. Moreover, the human fetal osteoblastic cell line exhibited extensive spreading and secreted elevated levels of collagen I and interleukin 1ßin the modified samples. Drug release evaluations demonstrated sustained CIP release through a diffusion mechanism, showcasing excellent antibacterial activity against pathogenic bacteria, includingStreptococcus mutans, Pseudomonas aeruginosa, andEscherichia coli.
Assuntos
Antibacterianos , Cerâmica , Lasers , Osteoblastos , Propriedades de Superfície , Zircônio , Zircônio/química , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Humanos , Antibacterianos/farmacologia , Antibacterianos/química , Cerâmica/química , Cerâmica/farmacologia , Linhagem Celular , Implantes Dentários/microbiologia , Fusobacterium nucleatum/efeitos dos fármacos , Teste de Materiais , Ciprofloxacina/farmacologia , Ciprofloxacina/química , Interleucina-1beta/metabolismo , Aderência Bacteriana/efeitos dos fármacos , Difração de Raios X , Microscopia Eletrônica de Varredura , Fosfatase Alcalina/metabolismo , Microscopia de Força Atômica , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologiaRESUMO
Oral malodor still constitutes a major challenge worldwide. A strong effort is invested in eliminating volatile sulfur compound-producing oral bacteria through organic natural products such as essential oils. Fusobacterium nucleatum is a known volatile sulfur compound-producing bacteria that inspires oral malodor. The aim of the present study was to test the effect of lavender essential oil on the bacterium's ability to produce volatile sulfide compounds, the principal components of oral malodor. Lavender (Lavandula angustifolia) essential oil was extracted by hydrodistillation and analyzed using GC-MS. The minimal inhibitory concentration (MIC) of lavender essential oil on Fusobacterium nucleatum was determined in a previous trial. Fusobacterium nucleatum was incubated anaerobically in the presence of sub-MIC, MIC, and above MIC concentrations of lavender essential oil, as well as saline and chlorhexidine as negative and positive controls, respectively. Following incubation, volatile sulfur compound levels were measured using GC (Oralchroma), and bacterial cell membrane damage was studied using fluorescence microscopy. Chemical analysis of lavender essential oil yielded five main components, with camphor being the most abundant, accounting for nearly one-third of the total lavender essential oil volume. The MIC (4 µL/mL) of lavender essential oil reduced volatile sulfur compound secretion at a statistically significant level compared to the control (saline). Furthermore, the level of volatile sulfur compound production attributed to 1 MIC of lavender essential oil was in the range of the positive control chlorhexidine with no significant difference. When examining bacterial membrane damage, 2 MIC of lavender essential oil (i.e., 8 µL/mL) demonstrated the same, showing antibacterial membrane damage values comparative to chlorhexidine. Since lavender essential oil was found to be highly effective in hindering volatile sulfur compound production by Fusobacterium nucleatum through the induction of bacterial cell membrane damage, the results suggest that lavender essential oil may be a suitable alternative to conventional chemical-based anti-malodor agents.
Assuntos
Fusobacterium nucleatum , Halitose , Lavandula , Testes de Sensibilidade Microbiana , Óleos Voláteis , Óleos Voláteis/farmacologia , Óleos Voláteis/química , Fusobacterium nucleatum/efeitos dos fármacos , Fusobacterium nucleatum/metabolismo , Halitose/microbiologia , Halitose/tratamento farmacológico , Halitose/metabolismo , Lavandula/química , Sulfetos/farmacologia , Sulfetos/química , Humanos , Óleos de Plantas/farmacologia , Óleos de Plantas/química , Cromatografia Gasosa-Espectrometria de Massas , Compostos Orgânicos Voláteis/farmacologia , Compostos Orgânicos Voláteis/química , Antibacterianos/farmacologia , Antibacterianos/químicaRESUMO
Pleural empyema can lead to significant morbidity and mortality despite chest drainage and antibiotic treatment, necessitating novel and minimally invasive interventions. Fusobacterium nucleatum is an obligate anaerobe found in the human oral and gut microbiota. Advances in sequencing and puncture techniques have made it common to detect anaerobic bacteria in empyema cases. In this report, we describe the case of a 65-year-old man with hypertension who presented with a left-sided encapsulated pleural effusion. Initial fluid analysis using metagenomic next-generation sequencing (mNGS) revealed the presence of Fusobacterium nucleatum and Aspergillus chevalieri. Unfortunately, the patient experienced worsening pleural effusion despite drainage and antimicrobial therapy. Ultimately, successful treatment was achieved through intrapleural metronidazole therapy in conjunction with systemic antibiotics. The present case showed that intrapleural antibiotic therapy is a promising measure for pleural empyema.
Assuntos
Antibacterianos , Empiema Pleural , Fusobacterium nucleatum , Terapia de Salvação , Humanos , Masculino , Idoso , Empiema Pleural/tratamento farmacológico , Empiema Pleural/microbiologia , Antibacterianos/administração & dosagem , Antibacterianos/uso terapêutico , Fusobacterium nucleatum/efeitos dos fármacos , Fusobacterium nucleatum/isolamento & purificação , Fusobacterium nucleatum/genética , Infecções por Fusobacterium/tratamento farmacológico , Infecções por Fusobacterium/complicações , Infecções por Fusobacterium/microbiologia , Metronidazol/uso terapêutico , Metronidazol/administração & dosagem , Sequenciamento de Nucleotídeos em Larga Escala , Resultado do TratamentoRESUMO
Intratumor microbes have attracted great attention in cancer research due to its influence on the tumorigenesis, progression and metastasis of cancer. However, the therapeutic strategies targeting intratumoral microbes are still in their infancy. Specific microorganisms, such as Fusobacterium nucleatum (F. nucleatum), are abundant in various cancer and always result in the CRC progression and chemotherapy resistance. Here, a combined anticancer and antibacterial therapeutic strategy is proposed to deliver antitumor drug to the tumors containing intratumor microbiota by the antibacerial polymeric drug carriers. We construct oral tellurium-containing drug carriers using a complex of tellurium-containing polycarbonate with cisplatin (PTE@CDDP). The results show that the particle size of the prepared nanoparticles could be maintained at about 105 nm in the digestive system environment, which is in line with the optimal particle size of oral nanomedicine. In vitro mechanism study indicates that the tellurium-containing polymers are highly effective in killing F.nucleatum through a membrane disruption mechanism. The pharmacokinetic experiments confirmed that PTE@CDDP has the potential function of enhancing the oral bioavailability of cisplatin. Both in vitro and in vivo studies show that PTE@CDDP could inhibit intratumor F.nucleatum and lead to a reduction in cell proliferation and inflammation in the tumor site. Together, the study identifies that the CDDP-loaded tellurium-containing nanoparticles have great potential for treating the F.nucleatum-promoted colorectal cancer (CRC) by combining intratumor microbiota modulation and chemotherapy. The synergistic therapeutic strategy provide new insight into treating various cancers combined with bacterial infection. STATEMENT OF SIGNIFICANCE: The synthesized antibacterial polymer was first employed to remodel the intratumor microbes in tumor microenvironment (TME). Moreover, it was the first report of tellurium-containing polymers against F.nucleatum and employed for treatment of the CRC. A convenient oral dosage form of cisplatin (CDDP)-loaded tellurium-containing nanoparticles (PTE@CDDP) was adopted here, and the synergistic antibacterial/chemotherapy effect occurred. The PTE@CDDP could quickly and completely eliminate F.nucleatum in a safe dose. In the CRC model, PTE@CDDP effectively reversed the inflammation level and even restored the intestinal barrier damaged by F.nucleatum. The ultrasensitive ROS-responsiveness of PTE@CDDP triggered the fast oxidation and efficient drug release of CDDP and thus a highly efficient apoptosis of the tumors. Therefore, the tellurium-containing polymers are expected to serve as novel antibacterial agents in vivo and have great potential in the F.nucleatum-associated cancers. The achievements provided new insight into treating CRC and other cancers combined with bacterial infection.
Assuntos
Antibacterianos , Neoplasias Colorretais , Portadores de Fármacos , Cimento de Policarboxilato , Telúrio , Telúrio/química , Telúrio/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Antibacterianos/farmacologia , Antibacterianos/química , Animais , Cimento de Policarboxilato/química , Portadores de Fármacos/química , Humanos , Cisplatino/farmacologia , Fusobacterium nucleatum/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Sinergismo Farmacológico , Linhagem Celular Tumoral , Antineoplásicos/farmacologia , Antineoplásicos/química , Nanopartículas/química , MasculinoRESUMO
Titanium implants are subject to bacterial adhesion and peri-implantitis induction, and biosurfactants bring a new alternative to the fight against infections. This work aimed to produce and characterize the biosurfactant from Bacillus subtilis ATCC 19,659, its anti-adhesion and antimicrobial activity, and cell viability. Anti-adhesion studies were carried out against Streptococcus sanguinis, Staphylococcus aureus, Fusobacterium nucleatum, Aggregatibacter actinomycetemcomitans, Porphyromonas gingivalis, and Proteus mirabilis as the minimum inhibitory concentration and the minimum bactericidal concentration. Cell viability was measured against osteoblast and fibroblast cells. The biosurfactant was classified as lipopeptide, with critical micelle concentration at 40 µg mL- 1, and made the titanium surface less hydrophobic. The anti-adhesion effect was observed for Staphylococcus aureus and Streptococcus sanguinis with 54% growth inhibition and presented a minimum inhibitory concentration of 15.7 µg mL- 1 for Streptococcus sanguinis and Aggregatibacter actinomycetemcomitans. The lipopeptide had no cytotoxic effect and demonstrated high potential application against bacterial biofilms.
Assuntos
Aderência Bacteriana , Biofilmes , Implantes Dentários , Lipopeptídeos , Testes de Sensibilidade Microbiana , Titânio , Titânio/farmacologia , Titânio/química , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Aderência Bacteriana/efeitos dos fármacos , Implantes Dentários/microbiologia , Lipopeptídeos/farmacologia , Humanos , Antibacterianos/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/fisiologia , Bacillus subtilis/efeitos dos fármacos , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/fisiologia , Porphyromonas gingivalis/crescimento & desenvolvimento , Aggregatibacter actinomycetemcomitans/efeitos dos fármacos , Propriedades de Superfície , Fibroblastos/efeitos dos fármacos , Fusobacterium nucleatum/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Tensoativos/farmacologiaRESUMO
Fusobacterium nucleatum (F. nucleatum), an oral anaerobe, is prevalent in colorectal cancer and is closely related to increased cancer cell growth, metastasis, and poor treatment outcomes. Bacterial vaccines capable of selectively eliminating bacteria present a promising approach to targeting intratumor F. nucleatum, thereby enhancing cancer treatment. Although adjuvants have been employed to enhance the immune response, the vaccine's effectiveness is constrained by inadequate T-cell activation necessary for eradicating intracellular pathogens. In this study, we developed a minimalistic, biomimetic nanovaccine by integrating highly immunostimulatory adjuvant cholesterol-modified CpG oligonucleotides into the autologously derived F. nucleatum membranes. Compared to the traditional vaccines consisting of inactivated bacteria and Alum adjuvant, the nanovaccine coupled with bacterial membranes and adjuvants could remarkably improve multiple antigens and adjuvant co-delivery to dendritic cells, maximizing their ability to achieve effective antigen presentation and strong downstream immune progress. Notably, the nanovaccine exhibits outstanding selective prophylactic and therapeutic effects, eliminating F. nucleatum without affecting intratumoral and gut microbiota. It significantly enhances chemotherapy efficacy and reduces cancer metastasis in F. nucleatum-infected colorectal cancer. Overall, this work represents the rational application of bacterial nanovaccine and provides a blueprint for future development in enhancing the antitumor effect against bacterial-infected cancer.
Assuntos
Vacinas Bacterianas , Neoplasias Colorretais , Fusobacterium nucleatum , Fusobacterium nucleatum/efeitos dos fármacos , Fusobacterium nucleatum/imunologia , Animais , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Humanos , Camundongos , Oligodesoxirribonucleotídeos/farmacologia , Oligodesoxirribonucleotídeos/administração & dosagem , Oligodesoxirribonucleotídeos/uso terapêutico , Nanopartículas/química , Microbioma Gastrointestinal/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/efeitos dos fármacos , Colesterol , Feminino , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Vacinas Anticâncer/farmacologia , Adjuvantes de Vacinas/farmacologia , Adjuvantes de Vacinas/uso terapêutico , Adjuvantes de Vacinas/administração & dosagem , Linhagem Celular Tumoral , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/uso terapêutico , NanovacinasRESUMO
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.
Assuntos
Citocinas , Ensaio de Imunoadsorção Enzimática , Fusobacterium nucleatum , Halitose , Microscopia Eletrônica de Varredura , Phyllanthus emblica , Extratos Vegetais , Porphyromonas gingivalis , Reação em Cadeia da Polimerase em Tempo Real , Phyllanthus emblica/química , Halitose/tratamento farmacológico , Halitose/microbiologia , Humanos , Extratos Vegetais/farmacologia , Método Duplo-Cego , Fusobacterium nucleatum/efeitos dos fármacos , Porphyromonas gingivalis/efeitos dos fármacos , Feminino , Fatores de Tempo , Masculino , Resultado do Tratamento , Adulto , Adulto Jovem , Receptor 2 Toll-Like/efeitos dos fármacos , Frutas/química , Estatísticas não Paramétricas , Mucosa Bucal/efeitos dos fármacos , Mucosa Bucal/microbiologia , Análise de Variância , Compostos de Enxofre/farmacologia , Compostos de Enxofre/análiseRESUMO
Anti-angiogenesis has emerged a promising strategy against colorectal cancer (CRC). However, the efficacy of anti-angiogenic therapy is greatly compromised by the up-regulated autophagy levels resulting from the evolutionary resistance mechanism and the presence of Fusobacterium nucleatum (F. nucleatum) in CRC. Herein, we report a cationic polymer capable of blocking autophagic flux to deliver plasmid DNA (pDNA) encoding soluble FMS-like tyrosine kinase-1 (sFlt-1) for enhanced anti-angiogenic therapy against F. nucleatum-associated CRC. The autophagy-inhibiting cationic polymer, referred to as PNHCQ, is synthesized by conjugating hydroxychloroquine (HCQ) into 3,3'-diaminodipropylamine-pendant poly(ß-benzyl-L-aspartate) (PAsp(Nors)), which can be assembled and electrostatically interacted with sFlt-1 plasmid to form PNHCQ/sFlt-1 polyplexes. Hydrophobic HCQ modification not only boosts transfection efficiency but confers autophagy inhibition activity to the polymer. Hyaluronic acid (HA) coating is further introduced to afford PNHCQ/sFlt-1@HA for improved tumor targeting without compromising on transfection. Consequently, PNHCQ/sFlt-1@HA demonstrates significant anti-tumor efficacy in F. nucleatum-colocalized HT29 mouse xenograft model by simultaneously exerting anti-angiogenic effects through sFlt-1 expression and down-regulating autophagy levels exacerbated by F. nucleatum challenge. The combination of anti-angiogenic gene delivery and overall autophagy blockade effectively sensitizes CRC tumors to anti-angiogenesis, providing an innovative approach for enhanced anti-angiogenic therapy against F. nucleatum-resident CRC. STATEMENT OF SIGNIFICANCE: Up-regulated autophagy level within tumors is considered responsible for the impaired efficacy of clinic antiangiogenic therapy against CRC colonized with pathogenic F. nucleatum. To tackle this problem, an autophagy-inhibiting cationic polymer is developed to enable efficient intracellular delivery of plasmid DNA encoding soluble FMS-like tyrosine kinase-1 (sFlt-1) and enhance anti-angiogenic therapy against F. nucleatum-associated CRC. HA coating that can be degraded by tumor-enriching hyaluronidase is further introduced for improved tumor targeting without compromising transfection efficiency. The well-orchestrated polyplexes achieve considerable tumor accumulation, efficient in vivo transfection, and effectively reinforce the sensitivity of CRC to the sFlt-1-derived anti-angiogenic effects by significantly blocking overall autophagy flux exacerbated by F. nucleatum challenge, thus harvesting robust antitumor outcomes against F. nucleatum-resident CRC.
Assuntos
Autofagia , Neoplasias Colorretais , Fusobacterium nucleatum , Fusobacterium nucleatum/efeitos dos fármacos , Neoplasias Colorretais/patologia , Neoplasias Colorretais/tratamento farmacológico , Animais , Autofagia/efeitos dos fármacos , Humanos , Técnicas de Transferência de Genes , Camundongos Nus , Camundongos , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Terapia Genética/métodos , Camundongos Endogâmicos BALB C , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Plasmídeos , Inibidores da Angiogênese/farmacologia , Hidroxicloroquina/farmacologia , Infecções por Fusobacterium/tratamento farmacológico , Infecções por Fusobacterium/complicaçõesRESUMO
BACKGROUND: Periodontal disease is the leading cause of tooth loss, and an association between periodontal disease and non-oral systemic diseases has been shown. Formation of biofilm by periodontal pathogens such as Fusobacterium nucleatum, Porphyromonas gingivalis, and Streptococcus mutans and their resistance to antimicrobial agents are at the root of persistent and chronic bacterial infections. METHODS: The bactericidal effect of far-ultraviolet (F-UV) light irradiation at 222 nm on periodontal bacteria was assessed qualitatively and quantitatively. The effect of biofilm disruption by F-UV light on periodontal bacteria was examined by crystal violet staining, and the morphologic changes of the biofilm after F-UV irradiation were explored by confocal laser microscopy and scanning electron microscopy. We developed a thin fiber-type 222 nm F-UV irradiator and studied its safety and effect of reducing bacteria in rodent models. RESULTS: F-UV light at 222 nm had a bactericidal effect on F. nucleatum, P. gingivalis, and S. mutans. Irradiation with F-UV light reduced the biofilm formed by the bacteria and sterilized them from within. Confocal laser microscopy showed a clear reduction in biofilm thickness, and scanning electron microscopy confirmed disintegration of the biofilm architecture. F-UV irradiation was less damaging to DNA and less cytotoxic than deep-ultraviolet light, and it reduced bacterial counts on the tooth surface. CONCLUSION: F-UV irradiation has the potential to destroy biofilm and act as a bactericide against pathogenic bacteria in the biofilm.
Assuntos
Biofilmes , Fusobacterium nucleatum , Periodontite , Porphyromonas gingivalis , Streptococcus mutans , Raios Ultravioleta , Biofilmes/efeitos dos fármacos , Biofilmes/efeitos da radiação , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/efeitos da radiação , Fusobacterium nucleatum/efeitos dos fármacos , Fusobacterium nucleatum/efeitos da radiação , Animais , Streptococcus mutans/efeitos dos fármacos , Streptococcus mutans/efeitos da radiação , Streptococcus mutans/fisiologia , Periodontite/microbiologia , Microscopia Confocal , Microscopia Eletrônica de Varredura , Camundongos , Viabilidade Microbiana/efeitos da radiação , Viabilidade Microbiana/efeitos dos fármacos , HumanosRESUMO
Antimicrobial peptides (AMPs) are becoming next-generation alternative antibacterial agents because of the rapid increase in resistance in bacteria against existing antibiotics, which can also be attributed to the formation of resilient biofilms. However, their widespread use is limited because of their poor absorption, higher dosage requirements, and delayed onset of the bioactivity to elicit a desired response. Here we developed a short AMP that specifically targeted Fusobacterium nucleatum. We conjugated 23R to a statherin-derived peptide (SDP) through rational design; this conjugate binds to FomA, a major porin protein of F. nucleatum. The SDP-tagged 23R exhibited rapid and highly specific bactericidal efficacy against F. nucleatum. Further, IC50 values were in the nanomolar range, and they were 100-fold lower than those obtained with unconjugated 23R. In a human gut microbiota model, 0.1 nM SDP-23R achieved 99% clearance of F. nucleatum ATCC 25586 without markedly altering resident microbiota. Here we demonstrated that binding-peptide-coupled AMPs show increased killing efficacy and specificity for the target pathogen without affecting the resident microbiota.
Assuntos
Antibacterianos , Peptídeos Antimicrobianos , Fusobacterium nucleatum , Fusobacterium nucleatum/efeitos dos fármacos , Peptídeos Antimicrobianos/farmacologia , Peptídeos Antimicrobianos/química , Humanos , Antibacterianos/farmacologia , Antibacterianos/química , Testes de Sensibilidade Microbiana , Microbioma Gastrointestinal/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Porinas/metabolismo , Porinas/genética , Porinas/química , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas da Membrana Bacteriana Externa/genéticaRESUMO
INTRODUCTION: Peri-implantitis is an inflammatory process around dental implants that is characterised by bone loss that may jeopardize the long-term survival of osseo integrated dental implants. The aim of this study was to create a surface coating on titanium abutments that possesses cellular adhesion and anti-microbial properties as a post-implant placement strategy for patients at risk of peri-implantitis. MATERIALS AND METHODSMETHODS: Titanium alloy Grade V stubs were coated with gold particles and then subjected to ceramic conversion treatment (CCT) at 620 °C for 3, 8 and 80 h. The surface characteristics and chemistry were assessed using scanning electron microscopy (SEM), energy dispersive spectroscopy (EDS), and X-ray diffraction (XRD) analysis. The leaching profile was investigated by inductively coupled plasma mass spectroscopy (ICP-MS) for all groups after 7, 14 and 28 days in contact with distilled water. A scratch test was conducted to assess the adhesion of the gold coating to the underlying titanium discs. Two bacterial species (Staphylococcus aureus (SA) & Fusobacterium nucleatum (FN)) were used to assess the antibacterial behaviour of the coated discs using a direct attachment assay test. The potential changes in surface chemistry by the bacterial species were investigated by grazing angle XRD. RESULTS: The gold pre-coated titanium discs exhibited good stability of the coating especially after immersion in distilled water and after bacterial colonisation as evident by XRD analysis. Good surface adhesion of the coating was demonstrated for gold treated discs after scratch test analysis, especially titanium, following a 3-hour (3 H) ceramic conversion treatment. All coated discs exhibited significantly improved antimicrobial properties against both tested bacterial species compared to untreated titanium discs. CONCLUSIONS: Ceramic conversion treated titanium with a pre-deposited gold layer showed improved antimicrobial properties against both SA and FN species than untreated Ti-C discs. Scratch test analysis showed good adherence properties of the coated discs the oxide layer formed is firmly adherent to the underlying titanium substrate, suggesting that this approach may have clinical efficacy for coating implant abutments.
Assuntos
Cerâmica , Dente Suporte , Fusobacterium nucleatum , Ouro , Microscopia Eletrônica de Varredura , Staphylococcus aureus , Propriedades de Superfície , Titânio , Difração de Raios X , Titânio/química , Ouro/química , Fusobacterium nucleatum/efeitos dos fármacos , Cerâmica/química , Staphylococcus aureus/efeitos dos fármacos , Teste de Materiais , Espectrometria por Raios X , Peri-Implantite , Implantes Dentários/microbiologia , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Anti-Infecciosos/farmacologia , Anti-Infecciosos/química , Antibacterianos/farmacologia , Antibacterianos/químicaRESUMO
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.
Assuntos
Antibacterianos , Aderência Bacteriana , Periodontite , Porphyromonas gingivalis , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/crescimento & desenvolvimento , Porphyromonas gingivalis/fisiologia , Humanos , Periodontite/microbiologia , Antibacterianos/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Extratos Vegetais/farmacologia , Extratos Vegetais/química , Catequina/farmacologia , Fusobacterium nucleatum/efeitos dos fármacos , Fusobacterium nucleatum/crescimento & desenvolvimento , Fusobacterium nucleatum/fisiologia , Adulto , Prevotella nigrescens/efeitos dos fármacos , Prevotella nigrescens/fisiologia , Feminino , Infecções por Bacteroidaceae/microbiologia , Masculino , Aggregatibacter actinomycetemcomitans/efeitos dos fármacos , Aggregatibacter actinomycetemcomitans/crescimento & desenvolvimento , Aggregatibacter actinomycetemcomitans/fisiologiaRESUMO
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.
Assuntos
Antibacterianos , Regeneração Óssea , Quitosana , Cobre , Fusobacterium nucleatum , Nanopartículas , Periodontite , Quitosana/química , Quitosana/farmacologia , Periodontite/tratamento farmacológico , Periodontite/microbiologia , Periodontite/terapia , Periodontite/patologia , Animais , Antibacterianos/farmacologia , Antibacterianos/química , Regeneração Óssea/efeitos dos fármacos , Ratos , Cobre/química , Cobre/farmacologia , Fusobacterium nucleatum/efeitos dos fármacos , Nanopartículas/química , Ratos Sprague-Dawley , Masculino , Sulfatos/química , Sulfatos/farmacologia , Dióxido de Silício/química , Dióxido de Silício/farmacologia , Testes de Sensibilidade MicrobianaRESUMO
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.
Assuntos
Ácido Aminolevulínico , Fusobacterium nucleatum , Periodontite , Fotoquimioterapia , Fármacos Fotossensibilizantes , Fusobacterium nucleatum/efeitos dos fármacos , Ácido Aminolevulínico/farmacologia , Ácido Aminolevulínico/uso terapêutico , Humanos , Periodontite/microbiologia , Periodontite/tratamento farmacológico , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Adulto , Masculino , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Feminino , Microbiota/efeitos dos fármacosRESUMO
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.