Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Eur Cell Mater ; 41: 421-430, 2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33782938

RESUMO

Bacterial contamination is hard to avoid during dental implant surgery. Macrophages and their polarisation play a decisive role in bacterial colonisation and tissue integration on bacterially contaminated dental implants. The present study investigated the role of macrophages in stimulating tissue coverage overgrowth of contaminating oral bacteria on polished titanium (Ti-P) and acid-etched zirconium dioxide (ZrO2-MA) dental implant materials. Different co-culture models were employed to determine phagocytosis rates of Streptococcus mitis or Staphylococcus aureus contaminating a dental implant surface and the influence of contaminating bacteria and osteoblasts (U2OS) on macrophage polarisation. S. aureus was phagocytized in higher numbers than S. mitis in bi-cultures on smooth Ti-P surfaces. Contaminating S. mitis stimulated near full polarisation of macrophages from a non-Ym1-expressing- to a Ym1-expressing-phenotype on smooth Ti-P, but on ZrO2-MA both phenotypes occurred. In tri-cultures with U2OS-cells on smooth Ti-P, a larger percentage of macrophages remained in their non-Ym1-expressing, "fighting" M1-like phenotype to clear Ti-P surfaces from contaminating bacteria. On ZrO2-MA surfaces, more macrophages tended towards their "fix- and-repair" M2-like phenotype than on Ti-P surfaces. Surface coverage of smooth, bacterially contaminated Ti-P surfaces by U2OS-cells was more effectively stimulated by fighting, M1-like macrophages than on ZrO2-MA surfaces. Comprehensive guidelines are provided for the development of infection-resistant, dental implant materials, including bacteria, tissue and immune cells. These guidelines point to more promising results for clinical application of Ti-P as compared with ZrO2-MA.


Assuntos
Implantes Dentários/microbiologia , Macrófagos/fisiologia , Fagocitose/fisiologia , Animais , Biofilmes/crescimento & desenvolvimento , Humanos , Ativação de Macrófagos/fisiologia , Macrófagos/microbiologia , Camundongos , Osteoblastos/efeitos dos fármacos , Osteoblastos/microbiologia , Fagocitose/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Streptococcus mitis/crescimento & desenvolvimento , Propriedades de Superfície , Titânio/farmacologia , Zircônio/farmacologia
2.
Exp Cell Res ; 354(1): 57-64, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28341446

RESUMO

The double-stranded RNA-dependent kinase (PKR), which is activated by double stranded RNA, induces inflammation by regulating NF-κB signaling. The NLR family pyrin domain-containing 3 (NLRP3) inflammasome also modulates inflammation in response to infection. Porphyromonas gingivalis (P.gingivalis) is an oral bacterium which is implicated in the pathogenesis of periodontal diseases. We previously reported that PKR is a key modulator of bone metabolism and inflammation in the periodontal tissue. PKR was also reported to induce inflammation in response to microbes by regulating the NLRP3 inflammasome, suggesting that PKR could affect inflammation along with NLRP3 in periodontal diseases. In this study, we investigated the effects of PKR on NLRP3 expression and NF-κB activity in P. gingivalis infected osteoblasts. We first constructed a SNAP26b-tagged P.gingivalis (SNAP-P. g.) and traced its internalization into the cell. SNAP-P. g. increased the activity of PKR and NF-κB and also induced NLRP3 expression in osteoblasts. Inhibition of NF-κB attenuated SNAP-P. g.-induced NLRP3 expression. The knockdown of PKR using shRNA decreased both the activity of NF-κB and the expression of NLRP3 induced by SNAP-P.g.. We therefore concluded that in osteoblasts, P. gingivalis activated PKR, which in turn increased NLRP3 expression by activating NF-κB. Our results suggest that PKR modulates inflammation by regulating the expression of the NLRP3 inflammasome through the NF-κB pathway in periodontal diseases.


Assuntos
Inflamassomos/genética , Inflamação/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Receptores Acoplados a Proteínas G/genética , Fator de Transcrição RelA/biossíntese , Células 3T3 , Animais , Regulação da Expressão Gênica/genética , Humanos , Inflamação/microbiologia , Inflamação/patologia , Camundongos , NF-kappa B/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/biossíntese , Osteoblastos/metabolismo , Osteoblastos/microbiologia , Osteoblastos/patologia , Bolsa Periodontal/genética , Bolsa Periodontal/microbiologia , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/patogenicidade , Receptores Acoplados a Proteínas G/biossíntese , Transdução de Sinais/genética , Fator de Transcrição RelA/genética
3.
Biomacromolecules ; 18(11): 3776-3787, 2017 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-28974094

RESUMO

In addition to customizing shapes of metal bone substitutes for patients, the 3D printing technique can reduce the modulus of the substitutes through the design and manufacture of interconnected porous structures, achieving the modulus match between substitute and surrounding bone to improve implant longevity. However, the porous bone substitutes take more risks of postoperative infection due to its much larger surface area compared with the traditional casting solid bone substitute. Here, we prepared of gentamicin-loaded silk fibroin coatings on 3D-printed porous cobalt-chromium-molybdenum (CoCrMo) bone substitutes via electrophoretic deposition technique. Through optimization, relatively intact, continuous, homogeneous, and conformal coatings with a thickness of 2.30 ± 0.58 µm were deposited around the struts with few pore blocked. The porous metal structures exhibited no loss in mechanical properties after the anode galvanic corrosion in EPD process. The initial osteoblastic response on coatings was better than that on metal surface, including cell spreading, proliferation and cytotoxicity. Antibacterial efficacy experiments showed that the coatings had an antibacterial effect on both adherent and planktonic bacteria within 1 week. These results suggested that the beneficial properties of anode electrophoretic deposited silk fibroin coatings could be exploited to improve the biological functionality of porous structures made of medical metals.


Assuntos
Substitutos Ósseos/síntese química , Materiais Revestidos Biocompatíveis/síntese química , Eletroforese/métodos , Gentamicinas/síntese química , Próteses e Implantes/microbiologia , Substitutos Ósseos/química , Substitutos Ósseos/uso terapêutico , Cromo/química , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/uso terapêutico , Cobalto/química , Fibroínas/química , Fibroínas/uso terapêutico , Gentamicinas/química , Gentamicinas/uso terapêutico , Humanos , Molibdênio/química , Osteoblastos/efeitos dos fármacos , Osteoblastos/microbiologia , Porosidade , Impressão Tridimensional , Próteses e Implantes/efeitos adversos , Seda/química , Seda/uso terapêutico , Propriedades de Superfície
4.
Nanotechnology ; 26(31): 315101, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26180062

RESUMO

Ceramic scaffolds are widely studied in the bone tissue engineering field due to their potential in regenerative medicine. However, adhesion of microorganisms on biomaterials with subsequent formation of antibiotic-resistant biofilms is a critical factor in implant-related infections. Therefore, new strategies are needed to address this problem. In the present study, three-dimensional and interconnected porous granules of nanostructured hydroxyapatite (nanoHA) incorporated with different amounts of zinc oxide (ZnO) nanoparticles were produced using a simple polymer sponge replication method. As in vitro experiments, granules were exposed to Staphylococcus aureus and Staphylococcus epidermidis and, after 24 h, the planktonic and sessile populations were assessed. Cytocompatibility towards osteoblast-like cells (MG63 cell line) was also evaluated for a period of 1 and 3 days, through resazurin assay and imaging flow cytometry analysis. As in vivo experiments, nanoHA porous granules with and without ZnO nanoparticles were implanted into the subcutaneous tissue in rats and their inflammatory response after 3, 7 and 30 days was examined, as well as their antibacterial activity after 1 and 3 days of S. aureus inoculation. The developed composites proved to be especially effective at reducing bacterial activity in vitro and in vivo for a weight percentage of 2% ZnO, with a low cell growth inhibition in vitro and no differences in the connective tissue growth and inflammatory response in vivo. Altogether, these results suggest that nanoHA-ZnO porous granules have a great potential to be used in orthopaedic and dental applications as a template for bone regeneration and, simultaneously, to restrain biomaterial-associated infections.


Assuntos
Antibacterianos/farmacologia , Materiais Biocompatíveis/toxicidade , Nanopartículas/química , Nanopartículas/microbiologia , Nanoporos , Osteoblastos/microbiologia , Animais , Linhagem Celular , Durapatita/química , Humanos , Técnicas In Vitro , Masculino , Nanoporos/ultraestrutura , Osteoblastos/efeitos dos fármacos , Porosidade , Ratos , Ratos Wistar , Infecções Estafilocócicas , Óxido de Zinco/química , Óxido de Zinco/farmacologia
5.
Implant Dent ; 23(4): 426-33, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24776940

RESUMO

OBJECTIVES: To evaluate the effectiveness of 4 procedures to disinfect implant surfaces intentionally inoculated with bacteria and afterward to evaluate osteoblast viability to the disinfected implant surfaces. MATERIALS AND METHODS: Eighty-eight commercially pure Osseotite and Nanotite titanium implant discs were inoculated with Porphyromonas gingivalis. The implant surfaces were disinfected with EDTA, tetracycline, citric acid, or neodymium-doped yttrium aluminum garnet (Nd:YAG) laser. The implant discs were then placed in cultures of osteoblast cells. RESULTS: Osseotite implant discs were easier to disinfect compared with the Nanotite implant discs. Citric acid and tetracycline were the most effective solutions for the disinfection of P. gingivalis from the Osseotite implant discs. CONCLUSION: The Nanotite implant discs were the most difficult to disinfect, likely because of their chemical and physical properties. Citric acid and tetracycline were most effective for disinfecting the Osseotite implant discs, and further clinical research is needed to verify these effects in vivo. The Nd:YAG laser was the weakest disinfection method, and it is not recommended for disinfecting implant surfaces until its effectiveness is improved.


Assuntos
Implantes Dentários , Desinfecção , Osteoblastos/citologia , Divisão Celular , Linhagem Celular , Humanos , Nanotecnologia , Osteoblastos/microbiologia , Porphyromonas gingivalis/efeitos dos fármacos , Porphyromonas gingivalis/crescimento & desenvolvimento , Propriedades de Superfície
6.
BMC Oral Health ; 14: 89, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25027664

RESUMO

BACKGROUND: Porphyromonas gingivalis has been shown to invade osteoblasts and inhibit their differentiation and mineralization in vitro. However, it is unclear if P. gingivalis can invade osteoblasts in vivo and how this would affect alveolar osteoblast/osteoclast dynamics. This study aims to answer these questions using a periodontitis mouse model under repetitive P. gingivalis inoculations. METHODS: For 3-month-old BALB/cByJ female mice, 10(9) CFU of P. gingivalis were inoculated onto the gingival margin of maxillary molars 4 times at 2-day intervals. After 2 weeks, another 4 inoculations at 2-day intervals were applied. Calcein was injected 7 and 2 days before sacrificing animals to label the newly formed bone. Four weeks after final inoculation, mice were sacrificed and maxilla collected. Immunohistochemistry, micro-CT, and bone histomorphometry were performed on the specimens. Sham infection with only vehicle was the control. RESULTS: P. gingivalis was found to invade gingival epithelia, periodontal ligament fibroblasts, and alveolar osteoblasts. Micro-CT showed alveolar bone resorption and significant reduction of bone mineral density and content in the infected mice compared to the controls. Bone histomorphometry showed a decrease in osteoblasts, an increase in osteoclasts and bone resorption, and a surprisingly increased osteoblastic bone formation in the infected mice compared to the controls. CONCLUSIONS: P. gingivalis invades alveolar osteoblasts in the periodontitis mouse model and cause alveolar bone loss. Although P. gingivalis appears to suppress osteoblast pool and enhance osteoclastic bone resorption, the bone formation capacity is temporarily elevated in the infected mice, possibly via some anti-microbial compensational mechanisms.


Assuntos
Perda do Osso Alveolar/microbiologia , Infecções por Bacteroidaceae/fisiopatologia , Osteoblastos/microbiologia , Osteoclastos/microbiologia , Osteogênese/fisiologia , Periodontite/microbiologia , Porphyromonas gingivalis/fisiologia , Perda do Osso Alveolar/patologia , Processo Alveolar/microbiologia , Processo Alveolar/patologia , Animais , Densidade Óssea/fisiologia , Contagem de Células , Modelos Animais de Doenças , Epitélio/microbiologia , Feminino , Fibroblastos/microbiologia , Fluoresceínas , Corantes Fluorescentes , Gengiva/microbiologia , Maxila/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos , Ligamento Periodontal/microbiologia , Ligamento Periodontal/patologia , Microtomografia por Raio-X/métodos
7.
BMC Microbiol ; 13: 5, 2013 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-23305098

RESUMO

BACKGROUND: Chronic periodontitis is an infectious disease of the periodontium, which includes the gingival epithelium, periodontal ligament and alveolar bone. The signature clinical feature of periodontitis is resorption of alveolar bone and subsequent tooth loss. The Gram-negative oral anaerobe, Porphyromonas gingivalis, is strongly associated with periodontitis, and it has been shown previously that P. gingivalis is capable of invading osteoblasts in a dose- and time-dependent manner resulting in inhibition of osteoblast differentiation and mineralization in vitro. It is not yet clear which receptors and cytoskeletal components mediate the invasive process, nor how the signaling pathways and viability of osteoblasts are affected by bacterial internalization. This study aimed to investigate these issues using an in vitro model system involving the inoculation of P. gingivalis ATCC 33277 into primary osteoblast cultures. RESULTS: It was found that binding between P. gingivalis fimbriae and integrin α5ß1 on osteoblasts, and subsequent peripheral condensation of actin, are essential for entry of P. gingivalis into osteoblasts. The JNK pathway was activated in invaded osteoblasts, and apoptosis was induced by repeated infections. CONCLUSIONS: These observations indicate that P. gingivalis manipulates osteoblast function to promote its initial intracellular persistence by prolonging the host cell life span prior to its intercellular dissemination via host cell lysis. The identification of molecules critical to the interaction between P. gingivalis and osteoblasts will facilitate the development of new therapeutic strategies for the prevention of periodontal bone loss.


Assuntos
Actinas/metabolismo , Endocitose , Interações Hospedeiro-Patógeno , Integrina alfa5beta1/metabolismo , Sistema de Sinalização das MAP Quinases , Osteoblastos/microbiologia , Porphyromonas gingivalis/patogenicidade , Animais , Apoptose , Células Cultivadas , Fímbrias Bacterianas/metabolismo , Camundongos , Ligação Proteica
8.
BMC Infect Dis ; 13: 267, 2013 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-23738922

RESUMO

BACKGROUND: In the fields of traumatology and orthopaedics staphylococci are the most frequently isolated pathogens. Staphylococcus aureus and Staphylococcus epidermidis are known to be the major causative agents of osteomyelitis. The increasing number of multiresistant Staphylococcus aureus and resistant coagulase-negative staphylococci as a trigger of complicated osteomyelitis and implant-associated infections is a major problem. Antibiotic therapy fails in 20% of cases. Therefore the development of novel antibiotics becomes necessary. METHODS: This study analyses tigecyclin, the first antibiotic of the glycylines, as a potential therapy for osteomyelitis caused by multiresistant Staphylococcus aureus. Therefore its intracellular activity and the potential use in polymethylmetacrylate-bone cement are examined. The intracellular activity of tigecyclin is determined by a human osteoblast infection model. The investigation of the biomechanical characteristics is conducted concerning the ISO 5833-guidelines. RESULTS: Tigecyclin shows in vitro an intracellular activity that ranges between the antimicrobial activity of gentamicin and rifampicin. A significant negative effect on the biomechanical characteristics with an impaired stability is detected after adding tigecyclin to polymethylmetacrylate-bone cement with a percentage of 1.225% per weight. CONCLUSIONS: This study shows that tigecyclin might be a potent alternative for the systemic therapy of osteomyelitis and implant-associated infections whereas the local application has to be reconsidered individually.


Assuntos
Cimentos Ósseos/química , Espaço Intracelular/microbiologia , Modelos Biológicos , Osteomielite/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/efeitos dos fármacos , Antibacterianos/farmacocinética , Antibacterianos/farmacologia , Fenômenos Biomecânicos , Células Cultivadas , Humanos , Teste de Materiais , Minociclina/análogos & derivados , Minociclina/farmacocinética , Minociclina/farmacologia , Osteoblastos/metabolismo , Osteoblastos/microbiologia , Osteomielite/microbiologia , Infecções Relacionadas à Prótese , Tigeciclina
9.
J Cell Biochem ; 113(5): 1724-32, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22213029

RESUMO

The antigen I/II (AgI/II) protein is a major surface protein that mediates the attachment of Streptococcus mutans (S. mutans) to the saliva-coated pellicle. Numerous studies have investigated not only the mechanisms by which AgI/II signaling is transduced within cells, but have also attempted to use AgI/II-specific antibodies to treat dental caries and host immune responses. However, little information is available about the effects of AgI/II on basic cellular events in bone cells. In this study, we examined the effects of the His-tagged recombinant N-terminal half of the AgI/II protein (rAgI/II-N) generated from S. mutans GS-5 on the viability, proliferation, and cell cycle progression of primary calvarial osteoblasts. We also investigated the mechanisms involved in the rAgI/II-N-mediated survival of serum-starved osteoblasts. We found that rAgI/II treatment attenuated the serum deprivation-induced decrease in cell viability and proliferation of osteoblasts. rAgI/II-N also prevented the loss of mitochondrial membrane potential (MMP), alterations in levels of two key mitochondrial Bcl-2 family proteins, and the accumulation of numerous cells into the sub-G(1) phase that were observed in serum-starved osteoblasts. Pharmacological inhibitors of phosphoinositide 3-kinase (PI3K), but not of extracellular signal-regulated kinase or Ras, blocked the rAgI/II-N-mediated protection against serum deprivation-induced cell death. Additional experiments revealed that the integrin α5ß1-mediated PI3K pathway is required for rAgI/II-N-mediated Akt phosphorylation in osteoblasts. Collectively, these results suggest that rAgI/II-N induces survival signals in serum-starved osteoblasts through integrin-induced PI3K/Akt signaling pathways.


Assuntos
Antígenos de Bactérias/fisiologia , Sobrevivência Celular/imunologia , Osteoblastos/microbiologia , Osteoblastos/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Streptococcus mutans/imunologia , Animais , Antígenos de Bactérias/administração & dosagem , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/imunologia , Ciclo Celular , Proliferação de Células , Células Cultivadas , Meios de Cultura Livres de Soro , Interações Hospedeiro-Patógeno/imunologia , Camundongos , Mitocôndrias/metabolismo , Modelos Biológicos , Osteoblastos/imunologia , Osteoblastos/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia , Transdução de Sinais , Streptococcus mutans/patogenicidade , Estresse Fisiológico
10.
Clin Oral Investig ; 16(3): 879-87, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21584693

RESUMO

Enterococcus faecalis (E. faecalis) is a Gram-positive bacterium, mostly recovered from root-filled teeth with persistent periapical lesions. Bacterial contamination of root canals inevitably results in interaction between E. faecalis and periapical tissues during the dynamic process of periapical inflammation. This study investigated the impact of heat-inactivated endodontic E. faecalis on the proliferation and the differentiation of ovine osteoblast-like cells, in an attempt to elucidate its putative enhanced pathogenicity mechanisms. Therefore, two different concentrations of a heat-inactivated endodontic E. faecalis isolate (2 × 10(6) or 2 × 10(8) CFU/ml) were incubated with ovine osteoblast-like cells for 7 and 14 days, respectively. Cells without antigen served as control. The effects of antigen on cell growth were evaluated by a proliferation assay (EZ4U). Furthermore, the assessment of alkaline phosphatase (ALP) activity, calcium deposition, and osteocalcin (OCN) gene expression through quantitative real-time PCR determined the degree of osteogenic cell differentiation. Scanning electron microscopy (SEM) was also performed to detect alterations in cell morphology. Interestingly, although highly concentrated E. faecalis increased cellular reproduction after 14 days, ALP activity and OCN gene expression decreased in an antigen concentration-dependent and incubation time-independent way. SEM images revealed E. faecalis adhesion on cells, a fact that might contribute to its virulence. These results suggest that E. faecalis stimulated cell multiplication, whereas it likely restrained cell differentiation of ovine osteoblast-like cells. In conclusion, the presence of E. faecalis in root canals may negatively affect periapical new bone formation, and thus, the healing of periapical lesions.


Assuntos
Enterococcus faecalis/fisiologia , Osteoblastos/microbiologia , Periodontite Periapical/microbiologia , Virulência , Fosfatase Alcalina/biossíntese , Fosfatase Alcalina/genética , Animais , Antígenos de Bactérias/fisiologia , Aderência Bacteriana , Cálcio/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Contagem de Colônia Microbiana , DNA Bacteriano/análise , Cavidade Pulpar/microbiologia , Humanos , Análise dos Mínimos Quadrados , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteocalcina/biossíntese , Osteocalcina/genética , Carneiro Doméstico , Calcificação de Dente
11.
Nano Lett ; 10(1): 202-7, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19968257

RESUMO

Bacterial infections present an enormous problem causing human suffering and cost burdens to healthcare systems worldwide. Here we present novel tunable antibacterial coatings which completely inhibit bacterial colonization by Staphylococcus epidermidis but allow normal adhesion and spreading of osteoblastic cells. The coatings are based on amine plasma polymer films loaded with silver nanoparticles. The method of preparation allows flexible control over the amount of loaded silver nanoparticles and the rate of release of silver ions.


Assuntos
Anti-Infecciosos/farmacologia , Infecções Bacterianas/prevenção & controle , Nanopartículas Metálicas/química , Adesividade , Infecções Bacterianas/tratamento farmacológico , Adesão Celular , Técnicas de Cultura de Células , Humanos , Íons , Testes de Sensibilidade Microbiana , Nanopartículas , Nanotecnologia/métodos , Osteoblastos/citologia , Osteoblastos/microbiologia , Polímeros/química , Prata/química , Staphylococcus epidermidis/efeitos dos fármacos
12.
Int J Paediatr Dent ; 21(3): 192-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21199004

RESUMO

OBJECTIVES: Osteomyelitis is an inflammatory process accompanied by bone destruction that is caused by bacterial infection, with most child cases showing a haematogenous origin and metaphysis of the long bones. The aim of the present study was to characterize streptococcal strains isolated from the blood of a child diagnosed with osteomyelitis in a long bone and investigate the biological properties related to virulence of strains associated with osteomyelitis. METHODS: Blood isolate species were determined based on the 16S rRNA sequence. Next, the blood isolates were analysed for phagocytosis susceptibility by polymorphonuclear leukocytes, platelet aggregation, inhibitory effects on osteoblastic cells, and their properties of adhesion with cells, and compared to the reference strain Streptococcus mitis ATCC49456. RESULTS: The blood isolates were found to be a single clone (named SA1101), which was determined to be S. mitis. The phagocytosis susceptibility of SA1101 was significantly lower than that of ATCC49456, while its platelet aggregation rate was higher. Furthermore, SA1101 showed an inhibitory effect toward the growth of osteoblastic cells and had greater properties of adhesion to those cells as compared to ATCC49456. CONCLUSIONS: These results suggest that S. mitis SA1101 is a possible etiological agent and caused osteomyelitis in the present case.


Assuntos
Osteomielite/microbiologia , Infecções Estreptocócicas/microbiologia , Streptococcus mitis/patogenicidade , Células 3T3 , Animais , Aderência Bacteriana , Criança , Contagem de Colônia Microbiana , Impressões Digitais de DNA , DNA Bacteriano/genética , Placa Dentária/microbiologia , Feminino , Humanos , Camundongos , Osteoblastos/microbiologia , Osteomielite/sangue , Fagocitose , Infecções Estreptocócicas/sangue , Streptococcus mitis/isolamento & purificação
13.
J Immunol ; 181(12): 8711-8, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19050291

RESUMO

Osteoimmunolgy involves the interaction of the immune system with skeletal elements. This interaction can lead to the formation of osseous lesions. To investigate how the acquired immune response could contribute to osteolytic lesions, we injected the periodontal pathogen Porphyromonas gingivalis adjacent to calvarial bone with or without prior immunization against the bacterium. Activation of the acquired immune response increased osteoclastogenesis and decreased coupled bone formation. The latter was accompanied by an increase in nuclear translocation of the transcription factor FOXO1 in vivo, increased apoptosis of bone-lining cells measured by the TUNEL assay and number of activated caspase-3 positive cells and a decrease in bone lining cell density. Further studies were conducted with MC3T3 osteoblastic cells. Apoptosis and increased FOXO1 DNA binding activity were induced when a combination of cytokines was tested, IL-beta, TNF-alpha, and IFN-gamma. Knockdown of FOXO1 by small interfering RNA significantly reduced cytokine stimulated apoptosis, cleaved caspase-3/7 activity and decreased mRNA levels of the proapoptotic genes, TNF-alpha, FADD, and caspase-3, -8, and -9. These results indicate that activation of the acquired immunity by a periodontal pathogen reduces the coupling of bone formation and resorption. This may occur by enhancing bone lining cell apoptosis through a mechanism that involves increased FOXO1 activation. These studies give insight into inflammatory bone diseases such as periodontal disease and arthritis were the formation of lytic lesions occurs in conjunction with deficient bone formation and activation of an acquired immune response.


Assuntos
Infecções por Bacteroidaceae/imunologia , Reabsorção Óssea/imunologia , Imunidade Ativa , Osteólise/imunologia , Osteólise/microbiologia , Periodontite/imunologia , Periodontite/microbiologia , Porphyromonas gingivalis/imunologia , Células 3T3 , Animais , Apoptose/imunologia , Infecções por Bacteroidaceae/metabolismo , Infecções por Bacteroidaceae/patologia , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/fisiologia , Imunidade Ativa/genética , Camundongos , Osteoblastos/imunologia , Osteoblastos/microbiologia , Osteoblastos/patologia , Osteólise/metabolismo , Periodontite/patologia , Periósteo/imunologia , Periósteo/microbiologia , Periósteo/patologia , RNA Interferente Pequeno/genética
14.
J Biomed Mater Res B Appl Biomater ; 108(1): 30-37, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-30883023

RESUMO

Bacterial cellulose is one of the most promising polymers of recent years. Herein, we present a possibility of BC application as a carrier of gentamycin antibiotic for the treatment and prevention of bone infections. We have shown that BC saturated with gentamycin significantly reduces the level of biofilm-forming bone pathogens, namely Staphylococcus aureus and Pseudomonas aeruginosa, and displays very low cytotoxicity in vitro against osteoblast cell cultures. Another beneficial feature of our prototype dressing is prolonged release of gentamycin, which provides efficient protection from microbial contamination and subsequent infection. Moreover, it seems that bacterial cellulose (BC) alone without any antimicrobial added, may serve as a barrier by significantly hampering the ability of the pathogen to penetrate to the bone structure. Therefore, a gentamycin-saturated BC dressing may be considered as a possible alternative for gentamycin collagen sponge broadly used in clinical setting. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 108B:30-37, 2020.


Assuntos
Bandagens , Biofilmes/efeitos dos fármacos , Celulose/química , Fêmur/microbiologia , Gentamicinas , Osteoblastos/microbiologia , Pseudomonas aeruginosa/fisiologia , Staphylococcus aureus/fisiologia , Animais , Biofilmes/crescimento & desenvolvimento , Linhagem Celular , Fêmur/metabolismo , Gentamicinas/química , Gentamicinas/farmacocinética , Gentamicinas/farmacologia , Humanos , Osteoblastos/metabolismo , Ratos
15.
PLoS One ; 15(4): e0231276, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32275737

RESUMO

OBJECTIVES: Alloyed metallic nanoparticles of silver and copper are effective against intracellular infection. However, systemic toxicity may arise due to the non-specific delivery of the nanoparticles. In addressing the issue, this study deals with the targeting of silver-copper-boron (ACB) nanoparticles to infected osteoblasts, which could decrease systemic toxicity and form the basis of targeting specific markers expressed in bone infections. METHODS: ACB nanoparticles were synthesized and conjugated to the Cadherin-11 antibody (OBAb). The effect of targeting nanoparticles against extracellular and intracellular S. aureus was determined by enumeration of bacterial growth. The binding of the targeting nanoparticles to infected osteoblasts as well as the visualization of live/dead bacteria due to treatment was carried out using fluorescence microscopy. MTT assay was used to determine the viability of osteoblasts with different concentrations of the nanoparticles. RESULTS: The ACB nanoparticles conjugated to OBAb (ACB-OBAb) were effective against extracellular S. aureus. The ACB-OBAb nanoparticles showed a 1.32 log reduction of intracellular S. aureus at a concentration of 1mg/L. The ACB-OBAb nanoparticles were able to bind to the infected osteoblast and showed toxicity to osteoblasts at levels ≥20mg/L. Also, the percentage of silver, copper, and boron in the nanoparticles determined the effectiveness of their antibacterial activity. CONCLUSION: The ACB-OBAb nanoparticles were able to target the osteoblasts and demonstrated significant antibacterial activity against intracellular S. aureus. Targeting shows promise as a strategy to target specific markers expressed on infected osteoblasts for efficient nanoparticle delivery, and further animal studies are recommended to test its efficacy in vivo.


Assuntos
Ligas/farmacologia , Osso e Ossos/citologia , Boro/farmacologia , Cobre/farmacologia , Espaço Intracelular/microbiologia , Nanopartículas Metálicas/química , Prata/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Antibacterianos/farmacologia , Caderinas/imunologia , Linhagem Celular , Endocitose/efeitos dos fármacos , Humanos , Espaço Intracelular/efeitos dos fármacos , Nanopartículas Metálicas/ultraestrutura , Testes de Sensibilidade Microbiana , Osteoblastos/efeitos dos fármacos , Osteoblastos/microbiologia
16.
Int J Pharm ; 576: 118941, 2020 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-31881261

RESUMO

Implanted scaffold or bone substitute is a common method to treat bone defects. However, the possible bone infection caused by orthopaedic surgery has created a challenging clinical problem and generally invalidate bone repair and regeneration. In this study, a poly (ε-caprolactone) (PCL)/polyethylene glycol (PEG)/roxithromycin (ROX) composite scaffold was prepared via melt electrohydrodynamic (EHD) 3D printing. Fourier transform infrared spectroscopy (FTIR) spectroscopy was performed to verify the existence of PEG and ROX in the scaffolds. By water contact angle measurement, the addition of both PEG and ROX was found to improve the hydrophilicity of the scaffolds. By in vitro drug release assay, the PCL/PEG/ROX scaffolds showed an initial burst drug release and subsequent long-term sustained release behaviour, which is favourable for the prevention and treatment of bone infections. The antibacterial assays against E. coli and S. aureus demonstrated that the composite scaffold with ROX possessed effective antibacterial activity, especially for S. aureus, the main cause of bone infection. The immunostaining and MTT assay with human osteoblast-like cells (MG63) indicated that cells showed good viability and growth on the scaffolds. Therefore, the melt EHD 3D printed PCL/PEG/ROX scaffold could be a promising anti-infective implant for bone tissue engineering.


Assuntos
Anti-Infecciosos/química , Anti-Infecciosos/farmacologia , Doenças Ósseas Infecciosas/tratamento farmacológico , Poliésteres/química , Polietilenoglicóis/química , Roxitromicina/química , Roxitromicina/farmacologia , Doenças Ósseas Infecciosas/microbiologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/microbiologia , Linhagem Celular Tumoral , Escherichia coli/efeitos dos fármacos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Osteoblastos/efeitos dos fármacos , Osteoblastos/microbiologia , Osteogênese/efeitos dos fármacos , Porosidade , Impressão Tridimensional , Staphylococcus aureus/efeitos dos fármacos , Engenharia Tecidual/métodos , Alicerces Teciduais
17.
Int Endod J ; 42(4): 375-80, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19298221

RESUMO

AIM: To investigate the receptor activator of nuclear factor-kappa B (NF-kappaB) ligand (RANKL) in osteoblastic cells stimulated with inflammatory mediators. METHODOLOGY: The expression of RANKL in human osteoblastic cell line U2OS stimulated by pro-inflammatory cytokine interleukin (IL)-1alpha and black-pigmented bacteria Porphyromonas endodontalis was investigated by Western blot and enzyme-linked immunosorbent assay (ELISA). The significance of the results obtained from control and treated groups was statistically analysed by the paired Student's t-test. RESULTS: IL-1alpha was found to upregulate RANKL production in U2OS cells (P < 0.05). Investigations of the time dependence of RANKL expression in IL-1alpha-treated cells revealed a rapid accumulation of RANKL protein after 1 h of exposure; it remained elevated throughout the 24-h incubation period shown by Western blot and ELISA. In addition, P. endodontalis also increased RANKL expression in U2OS cells after 4-h incubation period demonstrated by Western blot and ELISA (P < 0.05). CONCLUSIONS: IL-1alpha and P. endodontalis may be involved in developing apical periodontitis through the stimulation of RANKL production.


Assuntos
Interleucina-1alfa/metabolismo , Osteoblastos/metabolismo , Periodontite Periapical/metabolismo , Porphyromonas endodontalis/química , Ligante RANK/biossíntese , Perda do Osso Alveolar/metabolismo , Western Blotting , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/farmacologia , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-1alfa/farmacologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/microbiologia , Periodontite Periapical/imunologia , Periodontite Periapical/microbiologia , Ligante RANK/análise , Regulação para Cima
18.
Braz Oral Res ; 33: e025, 2019 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-31038565

RESUMO

Recently, it has been suggested that the anti-inflammatory hormone ghrelin (GHRL) and its receptor GHS-R may play a pivotal role in periodontal health and diseases. However, their exact regulation and effects in periodontitis are not known. The aim of this in-vitro study was to investigate the effect of microbial and inflammatory insults on the GHS-R1a expression in human osteoblast-like cells. MG-63 cells were exposed to interleukin (IL)-1ß and Fusobacterium nucleatum in the presence and absence of GHRL for up to 2 d. Subsequently, gene expressions of GHS-R1a, inflammatory mediators and matrix metalloproteinase were analyzed by real-time PCR. GHS-R protein synthesis and NF-κB p65 nuclear translocation were assessed by immunocytochemistry and immunofluorescence microscopy, respectively. IL-1ß and F. nucleatum caused a significant upregulation of GHS-R1a expression and an increase in GHS-R1a protein. Pre-incubation with a MEK1/2 inhibitor diminished the IL-1ß-induced GHS-R1a upregulation. IL-1ß and F. nucleatum also enhanced the expressions of cyclooxygenase 2, CC-chemokine ligand 2, IL-6, IL-8, and matrix metalloproteinase 1, but these stimulatory effects were counteracted by GHRL. By contrast, the stimulatory actions of IL-1ß and F. nucleatum on the GHS-R1a expression were further enhanced by GHRL. Our study provides original evidence that IL-1ß and F. nucleatum regulate the GHS-R/GHRL system in osteoblast-like cells. Furthermore, we demonstrate for the first time that the proinflammatory and proteolytic actions of IL-1ß and F. nucleatum on osteoblast-like cells are inhibited by GHRL. Our study suggests that microbial and inflammatory insults upregulate GHS-R1a, which may represent a protective negative feedback mechanism in human bone.


Assuntos
Fusobacterium nucleatum/fisiologia , Interleucina-1beta/farmacologia , Osteoblastos/química , Receptores de Grelina/análise , Análise de Variância , Células Cultivadas , Humanos , Imuno-Histoquímica , Microscopia de Fluorescência , Osteoblastos/efeitos dos fármacos , Osteoblastos/microbiologia , Periodontite/microbiologia , Periodontite/patologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Grelina/fisiologia , Estatísticas não Paramétricas , Regulação para Cima/fisiologia
19.
ACS Appl Mater Interfaces ; 11(27): 24588-24597, 2019 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-31199619

RESUMO

Additively manufactured selective laser melted titanium (SLM-Ti) opens the possibility of tailored medical implants for patients. Despite orthopedic implant advancements, significant problems remain with regard to suboptimal osseointegration at the interface between the implant and the surrounding tissue. Here, we show that applying a nanodiamond (ND) coating onto SLM-Ti scaffolds provides an improved surface for mammalian cell growth while inhibiting colonization of Staphylococcus aureus bacteria. Owing to the simplicity of our methodology, the approach is suitable for coating SLM-Ti geometries. The ND coating achieved 32 and 29% increases in cell density of human dermal fibroblasts and osteoblasts, respectively, after 3 days of incubation compared with the uncoated SLM-Ti substratum. This increase in cell density complements an 88% reduction in S. aureus detected on the ND-coated SLM-Ti substrata. This study paves a way to create facile antifouling SLM-Ti structures for biomedical implants.


Assuntos
Antibacterianos , Materiais Revestidos Biocompatíveis , Fibroblastos , Implantes Experimentais/microbiologia , Nanodiamantes/química , Osteoblastos , Staphylococcus aureus/crescimento & desenvolvimento , Titânio/química , Antibacterianos/química , Antibacterianos/farmacologia , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Fibroblastos/metabolismo , Fibroblastos/microbiologia , Fibroblastos/patologia , Humanos , Osteoblastos/metabolismo , Osteoblastos/microbiologia , Osteoblastos/patologia
20.
Biomaterials ; 204: 25-35, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30875516

RESUMO

Despite extensive engineering of tissue-contacting biomedical devices to control healing, these devices remain susceptible to bacterial colonization, biofilm formation, and chronic infection. The threat of selecting for resistance genes largely precludes sustained antimicrobial elution as a wide-spread clinical solution. In response, self-defensive surfaces have been developed where antimicrobial is released only when and where there is a bacterial challenge. We explore a new self-defensive approach using anionic microgels into which small-molecule cationic antimicrobials are loaded by complexation. We identify conditions where antimicrobial remains sequestered within the microgels for periods as long as weeks. However, bacterial contact triggers release and leads to local bacterial killing. We speculate that the close proximity of bacteria alters the local thermodynamic environment and interferes with the microgel-antimicrobial complexation. The contact-transfer approach does not require bacterial metabolism but instead appears to be driven by differences between the microgels and the bacterial cell envelope where there is a high concentration of negative charge and hydrophobicity. Contact with metabolizing macrophages or osteoblasts is, however, insufficient to trigger antimicrobial release, indicating that contact transfer can be specific to bacteria and suggesting an avenue to biomedical device surfaces that can simultaneously promote healing and resist infection.


Assuntos
Anti-Infecciosos/farmacologia , Materiais Biocompatíveis/farmacologia , Escherichia coli/efeitos dos fármacos , Staphylococcus epidermidis/efeitos dos fármacos , Resinas Acrílicas/química , Animais , Comunicação Celular/efeitos dos fármacos , Colistina/farmacologia , Feto/citologia , Humanos , Camundongos , Viabilidade Microbiana/efeitos dos fármacos , Microgéis , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/microbiologia , Células RAW 264.7 , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA