Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Infect Immun ; 76(11): 5120-6, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18765718

RESUMO

Osteomyelitis is an inflammatory disease of the bone that is characterized by the presence of necrotic bone tissue and increased osteoclast activity. Staphylococcus aureus is responsible for approximately 80% of all cases of human osteomyelitis. While the disease is especially difficult to treat, the pathogenesis of S. aureus-induced osteomyelitis is poorly understood. Elucidating the molecular mechanisms by which S. aureus induces osteomyelitis could lead to a better understanding of the disease and its progression and development of new treatments. Osteoblasts can produce several soluble factors that serve to modulate the activity or formation of osteoclasts. Receptor activator of NF-kappaB ligand (RANK-L) and prostaglandin E(2) (PGE(2)) are two such molecules which can promote osteoclastogenesis and stimulate bone resorption. In addition, previous studies in our laboratory have shown that osteoblasts produce inflammatory cytokines, such as interleukin 6, following infection with S. aureus, which could induce COX-2 and in turn PGE(2), further modulating osteoclast recruitment and differentiation. Therefore, we hypothesized that following infection with S. aureus, osteoblasts will express increased levels of RANK-L and PGE(2). The results presented in this study provide evidence for the first time that RANK-L mRNA and protein and PGE(2) expression are upregulated in S. aureus-infected primary osteoblasts. In addition, through the use of the specific COX-2 inhibitor NS 398, we show that when PGE(2) production is inhibited, RANK-L production is decreased. These data suggest a mechanism whereby osteoblasts regulate the production of RANK-L during infection.


Assuntos
Dinoprostona/biossíntese , Osteoblastos/metabolismo , Osteoblastos/microbiologia , Ligante RANK/biossíntese , Infecções Estafilocócicas/metabolismo , Animais , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Camundongos , Osteomielite/metabolismo , Osteomielite/microbiologia , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Staphylococcus aureus
2.
FEMS Microbiol Lett ; 278(2): 185-92, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18070069

RESUMO

Staphylococcus aureus is the principal etiological agent of osteomyelitis (bone infection), which is characterized by a progressive inflammatory response resulting in extensive damage to bone tissue. Recent studies have demonstrated the ability of S. aureus to invade and persist inside osteoblasts (bone matrix-forming cells) and other eukaryotic cells. The presence of intracellular S. aureus in bone tissue may be relevant to the pathology of osteomyelitis, a disease often refractory to antibiotic treatment and subject to recurrence months and even years after apparently successful therapy. The present study examined the production of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) following S. aureus infection, and whether expression of the molecule was induced by those osteoblasts containing intracellular S. aureus. Results from this study suggest that osteoblasts containing intracellular S. aureus induce TRAIL expression in uninfected osteoblasts present in infected cultures.


Assuntos
Osteoblastos/metabolismo , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/fisiologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Osteoblastos/citologia , Osteoblastos/microbiologia , Plasmídeos/genética , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Transformação Genética
3.
J Orthop Res ; 24(1): 87-93, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16419973

RESUMO

UNLABELLED: Staphylococcus aureus is responsible for 80% of human osteomyelitis. It can invade and persist within osteoblasts. Antibiotic resistant strains of S. aureus make successful treatment of osteomyelitis difficult. NULL HYPOTHESIS: antibiotic sensitivities of S. aureus do not change after exposure to the osteoblast intracellular environment. Human and mouse osteoblast cultures were infected and S. aureus cells were allowed to invade. Following times 0, 12, 24, and 48 h ( +/- the addition of erythromycin, clindamycin, and rifampin at times 0 or 12 h), the osteoblasts were lysed and intracellular bacteria enumerated. Transmission electron microscopy was performed on extracellular and intracellular S. aureus cells. In mouse osteoblasts, administration of bacteriostatic antibiotics at time 0 prevented the increase in intracellular S. aureus. If the antibiotics were delayed 12 h, this did not occur. When rifampin (bactericidal) was introduced at time 0 to human and mouse osteoblasts, there was a significant decrease in number of intracellular S. aureus within osteoblasts compared to control. If rifampin was delayed 12 h, this did not occur. Significant time-dependent S. aureus structural changes were observed after exposure to the osteoblast intracellular environment. These studies demonstrate that once S. aureus is established intracellularly for 12 h, the bacteria are less sensitive to antibiotics capable of eukaryotic cell penetration (statistically significant). These antibiotic sensitivity changes could be due in part to the observed structural changes. This leads to the rejection of our null hypotheses that the antibiotic sensitivities of S. aureus are unaltered by their location.


Assuntos
Farmacorresistência Bacteriana , Osteoblastos/microbiologia , Osteomielite/tratamento farmacológico , Osteomielite/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/efeitos dos fármacos , Animais , Células Cultivadas , Eritromicina/uso terapêutico , Gentamicinas/farmacologia , Humanos , Camundongos , Microscopia Eletrônica de Transmissão , Rifampina/uso terapêutico , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/ultraestrutura , Fatores de Tempo
4.
Bone ; 37(4): 504-12, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16027056

RESUMO

Incidences of osteomyelitis caused by Staphylococcus aureus have increased dramatically in recent years, in part, due to the appearance of community-acquired antibiotic-resistant strains. Therefore, understanding the pathogenesis of this organism has become imperative. Recently, we have described the surprising ability of bone-forming osteoblasts to secrete a number of important immune mediators when exposed to S. aureus in vitro. In the present study, we provide the first evidence for the in vivo production of the pivotal inflammatory chemokine, monocyte chemoattractant protein-1 (MCP-1), by osteoblasts during S. aureus-associated bone infection. Quantitative real-time PCR was employed to determine levels of mRNA encoding MCP-1 in vivo using a mouse model that closely resembles the pathology of trauma-induced staphylococcal osteomyelitis. Expression of this inflammatory chemokine and osteoblast-specific markers was investigated by confocal laser scanning microscopy in bone tissue from organ cultures of neonatal mouse calvaria and from the in vivo mouse model. Furthermore, the clinical relevancy of these findings was investigated by performing similar studies on infected human bone tissue from patients with S. aureus-associated osteomyelitis. Here, we confirm that expression of mRNA encoding MCP-1 is elevated in bacterially infected murine bone tissue. Importantly, we show that these increases translate into marked elevations in the expression of MCP-1 protein that co-localizes with osteoblast markers in infected bone tissue. Such increases could not be attributed solely to mechanical damage as a similar response was observed in infected but otherwise undamaged organ cultures. Finally, we have demonstrated the in vivo production of MCP-1 by osteoblasts in bone specimens from patients with S. aureus-associated osteomyelitis. As such, these studies demonstrate that bacterial challenge of osteoblasts during bone diseases such as staphylococcal osteomyelitis induces cells to produce a key inflammatory chemokine that can direct appropriate host responses or may contribute to progressive inflammatory damage.


Assuntos
Quimiocina CCL2/biossíntese , Osteoblastos/metabolismo , Osteomielite/metabolismo , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus/isolamento & purificação , Animais , Animais Recém-Nascidos , Quimiocina CCL2/genética , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Osteomielite/microbiologia , RNA Mensageiro/genética , Infecções Estafilocócicas/microbiologia
5.
Bone ; 33(5): 812-21, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14623057

RESUMO

A growing body of evidence has shown that bacterially challenged osteoblasts can play a significant role in the initiation of inflammatory immune responses at sites of bone disease. We have recently demonstrated the surprising ability of osteoblasts exposed to bacteria to express CD40, a molecule that plays a critical costimulatory role in the activation of T lymphocytes. In the present study, we have extended our investigations into the ability of osteoblasts to interact with CD4+ T lymphocytes by determining the expression of antigen-presenting major histocompatibility complex (MHC) class II molecules in murine and human osteoblasts following exposure to two common pathogens of bone, Staphylococcus aureus and Salmonella. Cultured osteoblasts were found to respond rapidly to bacterial challenge by induction of mRNA encoding MHC class II molecules or its transcriptional regulator. Increased mRNA expression translated into expression of MHC class II proteins in murine and human osteoblasts as determined by Western blot analysis and by immunohistochemical and immunofluorescent microscopy. Furthermore, the increased surface expression of these molecules on osteoblasts exposed to bacteria was confirmed by FACS analysis. Finally, we show that bacterial challenge results in the elevated functional expression of MHC class II molecules on osteoblasts by demonstrating the enhanced ability of these cells to interact with T lymphocytes and to initiate antigen-specific T cell activation. Taken together, these data suggest a previously unappreciated role for osteoblasts in the initiation of T lymphocyte activation at sites of bacterial infection in bone tissue.


Assuntos
Infecções Bacterianas/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Genes MHC da Classe II/fisiologia , Osteoblastos/metabolismo , Animais , Células Cultivadas , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Salmonella enterica/genética , Salmonella enterica/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo
6.
BMC Microbiol ; 3: 5, 2003 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-12697056

RESUMO

BACKGROUND: Staphylococcus aureus infection of normal osteoblasts induces expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). RESULTS: Normal osteoblasts were incubated in the presence of purified bacterial products over a range of concentrations. Results demonstrate that purified surface structures and a selected superantigen present in the extracellular environment are not capable of inducing TRAIL expression by osteoblasts. Osteoblasts were co-cultured with S. aureus at various multiplicities of infection utilizing cell culture chamber inserts. Results of those experiments suggest that direct contact between bacteria and osteoblasts is necessary for optimal TRAIL induction. Finally, S. aureus infection of osteoblasts in the presence of anti-TRAIL antibody demonstrates that TRAIL mediates caspase-8 activation and apoptosis of infected cells. CONCLUSIONS: Collectively, these findings suggest a mechanism whereby S. aureus mediates bone destruction via induction of osteoblast apoptosis.


Assuntos
Apoptose , Caspases/metabolismo , Glicoproteínas de Membrana/biossíntese , Osteoblastos/microbiologia , Staphylococcus aureus/fisiologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Caspase 8 , Células Cultivadas , Ativação Enzimática , Humanos , Ligantes , Glicoproteínas de Membrana/fisiologia , Camundongos , Osteoblastos/enzimologia , Osteoblastos/metabolismo , Osteoblastos/patologia , Staphylococcus aureus/química , Ligante Indutor de Apoptose Relacionado a TNF , Fator de Necrose Tumoral alfa/fisiologia
7.
J Biomed Mater Res A ; 95(2): 574-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20725968

RESUMO

Titanium alloys (Ti) are the preferred material for orthopedic applications. However, very often, these metallic implants loosen over a long period and mandate revision surgery. For implant success, osteoblasts must adhere to the implant surface and deposit a mineralized extracellular matrix (ECM). Here, we utilized UV-killed Staphylococcus aureus as a novel osteoconductive coating for Ti surfaces. S. aureus expresses surface adhesins capable of binding to bone and biomaterials directly. Furthermore, interaction of S. aureus with osteoblasts activates growth factor-related pathways that potentiate osteogenesis. Although UV-killed S. aureus cells retain their bone-adhesive ability, they do not stimulate significant immune modulator expression. All of the abovementioned properties were utilized for a novel implant coating so as to promote osteoblast recruitment and subsequent cell functions on the bone-implant interface. In this study, osteoblast adhesion, proliferation, and mineralized ECM synthesis were measured on Ti surfaces coated with fibronectin with and without UV-killed bacteria. Osteoblast adhesion was enhanced on Ti alloy surfaces coated with bacteria compared to uncoated surfaces, while cell proliferation was sustained comparably on both surfaces. Osteoblast markers such as collagen, osteocalcin, alkaline phosphatase activity, and mineralized nodule formation were increased on Ti alloy coated with bacteria compared to uncoated surfaces.


Assuntos
Materiais Biocompatíveis/metabolismo , Regeneração Óssea , Osso e Ossos , Adesão Celular/fisiologia , Diferenciação Celular/fisiologia , Osteoblastos/fisiologia , Staphylococcus aureus/efeitos da radiação , Animais , Materiais Biocompatíveis/química , Osso e Ossos/química , Osso e Ossos/metabolismo , Células Cultivadas , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/metabolismo , Humanos , Implantes Experimentais , Teste de Materiais , Camundongos , Osteoblastos/citologia , Staphylococcus aureus/citologia , Propriedades de Superfície , Titânio/química , Raios Ultravioleta
8.
Biomed Mater ; 3(3): 034114, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18708713

RESUMO

The goal of this investigation is to develop poly(DL-lactide-co-glycolide) (PLGA) nanoparticles for the delivery of antibiotics such as nafcillin to osteoblasts. This is important in order to treat Staphylococcus aureus-mediated osteomyelitis. The latter is often chronic and highly resistant to antibiotics. Nafcillin (a penicillinase-resistant penicillin)-loaded nanoparticles were prepared by a single emulsion/solvent evaporation method. In vitro drug release studies were conducted in an incubator shaker at 37 degrees C in phosphate buffer saline. Drug loading and release were determined by UV-Vis spectroscopy. A viability study was conducted in S. aureus-infected mouse osteoblasts. In vitro release study showed an initial burst release and a second phase of slow release. Following 24 and 48 h of incubation, all formulations of nanoparticles loaded with nafcillin either killed or significantly reduced all of the intracellular bacteria. Our data demonstrate that effective killing of intracellular S. aureus is possible by treating the infected osteoblasts with nanoparticles loaded with nafcillin.


Assuntos
Portadores de Fármacos/química , Ácido Láctico/química , Nafcilina/administração & dosagem , Nanopartículas/química , Osteoblastos/efeitos dos fármacos , Osteoblastos/microbiologia , Ácido Poliglicólico/química , Staphylococcus aureus/efeitos dos fármacos , Animais , Antibacterianos/administração & dosagem , Antibacterianos/química , Células Cultivadas , Portadores de Fármacos/administração & dosagem , Humanos , Camundongos , Nafcilina/química , Nanopartículas/administração & dosagem , Nanopartículas/ultraestrutura , Osteomielite/tratamento farmacológico , Tamanho da Partícula , Copolímero de Ácido Poliláctico e Ácido Poliglicólico
9.
Infect Immun ; 71(3): 1209-16, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12595434

RESUMO

Bacterially induced bone infections often result in significant local inflammatory responses which are coupled with loss of bone. However, the mechanisms necessary for the protective host response, or those responsible for pathogen-induced bone loss, are not clear. Recent evidence demonstrates that bacterially infected osteoblasts secrete chemokines and cytokines, suggesting that these cells may have an unappreciated role in supporting localized inflammation. In this study, mouse and human osteoblasts were investigated for their ability to express functional CD40 upon exposure to two important pathogens of bone, Staphylococcus aureus and Salmonella enterica serovar Dublin. Bacterial infection of cultured mouse or human osteoblasts resulted in increased CD40 mRNA and CD40 protein expression induced by either pathogen. Importantly, CD40 expression by osteoblasts was functional, as assessed by ligation of this molecule with recombinant, soluble CD154. CD40 activity was assessed by induction of interleukin-6 and granulocyte-macrophage colony-stimulating factor in osteoblasts following ligation. Cocultures of activated CD4(+) T lymphocytes and osteoblasts could interact via CD40 and CD154, since an antibody against CD40 could block macrophage inflammatory protein-1alpha secretion. Taken together, these studies conclusively demonstrate that infected osteoblasts can upregulate expression of functional CD40 molecules which mediate cytokine secretion. This surprising result further supports the notion that bone-forming osteoblasts can directly interact with CD154-expressing cells (i.e., T lymphocytes) and can contribute to the host response during bone infection.


Assuntos
Antígenos CD40/análise , Osteoblastos/metabolismo , Osteoblastos/microbiologia , Salmonella enterica/patogenicidade , Staphylococcus aureus/patogenicidade , Animais , Linfócitos T CD4-Positivos/metabolismo , Antígenos CD40/genética , Antígenos CD40/fisiologia , Ligante de CD40/fisiologia , Quimiocina CCL4 , Citocinas/biossíntese , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Proteínas Inflamatórias de Macrófagos/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Osteoblastos/química , RNA Mensageiro
10.
Am J Pathol ; 164(4): 1399-406, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15039227

RESUMO

Staphylococcus aureus is the single most common cause of osteomyelitis in humans. Incidences of osteomyelitis caused by S. aureus have increased dramatically in recent years, in part due to the appearance of community-acquired antibiotic resistant strains. Therefore, understanding the pathogenesis of this organism has become imperative. Recently, we have described the surprising ability of bone-forming osteoblasts to secrete a number of important immune mediators when exposed to S. aureus in vitro. In the present study, we provide the first evidence for the in vivo production of such molecules by osteoblasts during bacterial infection of bone. These studies demonstrate the expression of the key inflammatory cytokine interleukin-6 by osteoblasts in organ cultures of neonatal mouse calvaria, and in vivo using a mouse model that closely resembles the pathology of trauma-induced staphylococcal osteomyelitis, as determined by confocal microscopic analysis. Importantly, we have established the clinical relevancy of these findings in infected human bone tissue from patients with S. aureus-associated osteomyelitis. As such, these studies demonstrate that bacterial challenge of osteoblasts during bone diseases, such as osteomyelitis, induces cells to produce inflammatory molecules that can direct appropriate host responses or contribute to progressive inflammatory damage.


Assuntos
Interleucina-6/biossíntese , Osteoblastos/imunologia , Osteomielite/metabolismo , Infecções Estafilocócicas/imunologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Camundongos , Microscopia Confocal , Técnicas de Cultura de Órgãos , Osteomielite/etiologia , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Crânio/fisiologia , Infecções Estafilocócicas/complicações , Staphylococcus aureus/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA