Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Orthop Res ; 38(4): 861-870, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31692074

RESUMO

While Gram-positive organisms are the most common causative agent of initial bone infections, the percentage of Gram-negative species increases in reoccurring bone infections. As bacterial internalization has been suggested as one cause of reoccurring bone infection, we tested the hypothesis that Gram-negative species of bacteria can be internalized into bone cells. Using the MLO-A5 and the MLO-Y4 cell lines as our cell models, we demonstrated that the Gram-negative species, Proteus mirabilis and Serratia marcescens, can be internalized in these cells using an internalization assay. This rate at which these two species were internalized was both time- and initial concentration-dependent. Confocal analysis demonstrated the presence of internalized bacteria within both cell types. Inhibition of the cellular uptake with methyl-ß-cyclodextrin and chloroquine both reduced internalized bacteria, indicating that this process is, at least in part, cell mediated. Finally, we demonstrated that the presence of internalized P. mirabilis did not impact cell viability, measured either by lactate dehydrogenase (LDH) release or 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) activity, while the presence of S. marcescens, on the other hand, both increased LDH release and reduced MTT activity, indicating a loss of cell viability in response to the organism. These results indicated that both species of Gram-negative bacteria can be internalized by bone cells and that these internalized bacteria could potentially result in reoccurring bone infections. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:861-870, 2020.


Assuntos
Interações Hospedeiro-Patógeno , Osteócitos/microbiologia , Proteus mirabilis/fisiologia , Serratia marcescens/fisiologia , Animais , Linhagem Celular , Endocitose , Camundongos , Microscopia Confocal
2.
Angew Chem Int Ed Engl ; 55(42): 13240-13243, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27709816

RESUMO

A tandem allene aziridination/[4+3]/reduction sequence converts simple homoallenic sulfamates into densely functionalized aminated cycloheptenes, where the relative stereochemistry at five contiguous asymmetric centers can be controlled through the choice of the solvent and the reductant. The products resulting from this chemistry can be readily transformed into complex molecular scaffolds which contain up to seven contiguous stereocenters.


Assuntos
Alcenos/química , Aziridinas/química , Cicloeptanos/síntese química , Ciclização , Cicloeptanos/química , Conformação Molecular , Oxirredução , Estereoisomerismo
3.
Org Lett ; 18(2): 284-7, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26741730

RESUMO

Oxidative allene amination provides rapid access to densely functionalized amine-containing stereotriads through highly reactive bicyclic methyleneaziridine intermediates. This strategy has been demonstrated as a viable approach for the construction of the densely functionalized aminocyclitol core of jogyamycin, a natural product with potent antiprotozoal activity. Importantly, the flexibility of oxidative allene amination will enable the syntheses of modified aminocyclitol analogues of the jogyamycin core.


Assuntos
Pactamicina/análogos & derivados , Pactamicina/síntese química , Alcadienos/química , Aminação , Aminas/química , Estrutura Molecular , Oxirredução , Pactamicina/química , Pactamicina/farmacologia , Estereoisomerismo , Streptomyces/química
4.
J Biomed Mater Res B Appl Biomater ; 103(7): 1381-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25389082

RESUMO

As peri-prosthetic infection is one of the most devastating complications associated with implant placement, we have reasoned that such infection can be largely subverted by development of antibacterial implants. Our previous work demonstrated that covalent coupling of vancomycin to titanium alloy prevented colonization by the Gram-positive pathogens, Staphylococcus aureus and Staphylococcus epidermidis. Some orthopedic devices, including permanent prosthesis anchors, and most dental implants are transcutaneous or transmucosal and can be prone to colonization by Gram-negative pathogens. We report here the successful covalent coupling of the broad-spectrum antibiotic, tetracycline (TET), to titanium surfaces (Ti-TET) to retard Gram-negative colonization. Synthetic progress was followed by changes in water contact angle, while the presence of TET was confirmed by immunofluorescence. Ti-TET actively prevented colonization in the presence of bathing Escherichia coli, both by fluorescence microscopy and direct counting. Finally, the Ti-TET surface supported osteoblastic cell adhesion and proliferation over a 72-h period. Thus, this new surface offers a powerful means to protect transcutaneous implants from adhesion of Gram-negative pathogens, decreasing the need for replacement of this hardware.


Assuntos
Aderência Bacteriana/efeitos dos fármacos , Materiais Revestidos Biocompatíveis , Escherichia coli/crescimento & desenvolvimento , Osteoblastos/metabolismo , Tetraciclina , Titânio , Linhagem Celular Transformada , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Humanos , Tetraciclina/química , Tetraciclina/farmacologia , Titânio/química , Titânio/farmacologia
5.
J Am Chem Soc ; 134(26): 10807-10, 2012 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-22708990

RESUMO

Nitrogen-containing stereotriads, compounds with three adjacent stereodefined carbons, are commonly found in biologically important molecules. However, the preparation of molecules bearing these motifs can be challenging. Herein, we describe a modular oxidation protocol which converts a substituted allene to a triply functionalized amine of the form C-X/C-N/C-Y. The key step employs a Rh-catalyzed intramolecular conversion of the allene to a strained bicyclic methylene aziridine. This reactive intermediate is further elaborated to the target products, often in one reaction vessel and with effective transfer of the axial chirality of the allene to point chirality in the stereotriad.


Assuntos
Alcenos/química , Aminação , Aziridinas/síntese química , Catálise , Compostos Organometálicos/química , Oxirredução , Ródio/química , Estereoisomerismo , Ácidos Sulfônicos/química
6.
Cells Tissues Organs ; 194(2-4): 274-8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21597271

RESUMO

The goal of this investigation was to ascertain whether bone cells undergo autophagy and to determine if this process is regulated by environmental factors. We showed that osteocytes in both murine and human cortical bone display a punctuate distribution of microtubule-associated protein light chain 3, indicative of autophagy. In addition, we noted a basal level of autophagy in preosteocyte-like murine long bone-derived osteocytic (MLO)-A5 cells. Autophagy was upregulated following nutrient deprivation and hypoxic culture, stress conditions that osteocytes encounter in vivo. Furthermore, in response to calcium stress, the transcription factor hypoxia inducible factor 1 regulated MLO-A5 autophagy. Finally, we showed that the more differentiated MLO-Y4 osteocyte-like cells exhibited a significant basal autophagic flux. Based on these findings, we suggest that raising the level of autophagic flux is a mechanism by which differentiated bone cells survive in a stressful environment.


Assuntos
Autofagia , Osso e Ossos/citologia , Meio Ambiente , Osteócitos/citologia , Animais , Autofagia/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Osso e Ossos/ultraestrutura , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Osteócitos/efeitos dos fármacos , Osteócitos/metabolismo , Osteócitos/ultraestrutura , Fenótipo , Ratos , Ratos Wistar , Tapsigargina/farmacologia
7.
Bone ; 48(3): 631-8, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21035576

RESUMO

Bacterial contamination of bone allograft is a significant complication of orthopedic surgery. To address this issue, we have engineered a method for covalently modifying bone allograft tissue with the antibiotic vancomycin. The goal of this investigation was to compare the biocidal properties of this new allograft material with those of vancomycin physisorbed onto graft material. The duration of antibiotic release from the vancomycin-modified allograft matrix was determined, and no elution was observed. In contrast, the adsorbed antibiotic showed a peak elution at 24h that then decreased over several days. We next used an Staphylococcus aureus disk diffusion assay to measure the activity of the eluted vancomycin. Again we found that no active antibiotic was eluted from the covalently modified allograft. Similarly, when the vancomycin-modified allograft morsel was used in the assay, no measurable elution was observed; amounts of antibiotic released from the adsorbed samples inhibited S. aureus growth for 4-7 days. Probably the most telling property of the allograft was that after 2 weeks, the tethered allograft was able to resist bacterial colonization. Unlike the elution system in which vancomycin was depleted over the course of days-weeks, the antibiotic on the allograft was stably bound even after 300 days, while its biocidal activity remained undiminished for 60 days. This finding was in stark contrast to the antibiotic impregnated allograft, which was readily colonized by bacteria. Finally we chose to evaluate three indicators of cell function: expression of a key transcription factor, expression of selected transcripts, and assessment of cell morphology. Since the tethered antibiotic appeared to have little or no effect on any of these activities, it was concluded that the stable, tethered antibiotic prevented bacterial infection while not modifying bone cell function.


Assuntos
Antibacterianos/farmacologia , Transplante Ósseo , Vancomicina/farmacologia , Adsorção/efeitos dos fármacos , Antibacterianos/química , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Contagem de Colônia Microbiana , Estabilidade de Medicamentos , Feto/citologia , Fluorescência , Perfilação da Expressão Gênica , Humanos , Testes de Sensibilidade Microbiana , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Fenótipo , Fatores de Tempo , Transplante Homólogo , Vancomicina/química
8.
Antimicrob Agents Chemother ; 55(2): 487-94, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21098245

RESUMO

Infection is an important medical problem associated with the use of bone allografts. To retard bacterial colonization, we have recently reported on the modification of bone allografts with the antibiotic vancomycin (VAN). In this report, we examine the ability of this antibiotic-modified allograft to resist bacterial colonization and biofilm formation. When antibiotic was coupled to the allograft, a uniform distribution of the antibiotic was apparent. Following challenges with Staphylococcus aureus for 6 h, the covalently bonded VAN decreased colonization as a function of inoculum, ranging from 0.8 to 2.0 log(10) CFU. Furthermore, the VAN-modified surface resisted biofilm formation, even in topographical niches that provide a protected environment for bacterial adhesion. Attachment of the antibiotic to the allograft surface was robust, and the bonded VAN was stable whether incubated in aqueous media or in air, maintaining levels of 75 to 100% of initial levels over 60 days. While the VAN-modified allograft inhibited the Gram-positive S. aureus colonization, in keeping with VAN's spectrum of activity, the VAN-modified allograft was readily colonized by the Gram-negative Escherichia coli. Finally, initial toxicity measures indicated that the VAN-modified allograft did not influence osteoblast colonization or viability. Since the covalently tethered antibiotic is stable, is active, retains its specificity, and does not exhibit toxicity, it is concluded that this modified allograft holds great promise for decreasing bone graft-associated infections.


Assuntos
Antibacterianos/farmacologia , Transplante Ósseo , Osso e Ossos/química , Osso e Ossos/microbiologia , Staphylococcus aureus/crescimento & desenvolvimento , Vancomicina/farmacologia , Antibacterianos/química , Aderência Bacteriana/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Contagem de Colônia Microbiana , Humanos , Microscopia Confocal , Osteoblastos/citologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/fisiologia , Vancomicina/química
9.
Am J Physiol Cell Physiol ; 299(5): C922-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20660162

RESUMO

Whereas recent work has demonstrated the role of oxygen tension in the regulation of skeletal cell function and viability, the microenvironmental oxemic status of bone cells remains unknown. In this study, we have employed the Krogh cylinder model of oxygen diffusion to predict the oxygen distribution profiles in cortical and cancellous bone. Under the assumption of saturation-type Michaelis-Menten kinetics, our numerical modeling has indicated that, under steady-state conditions, there would be oxygen gradients across mature osteons and trabeculae. In Haversian bone, the calculated oxygen tension decrement ranges from 15 to 60%. For trabecular bone, a much shallower gradient is predicted. We note that, in Haversian bone, the gradient is largely dependent on osteocyte oxygen utilization and tissue oxygen diffusivity; in trabecular bone, the gradient is dependent on oxygen utilization by cells lining the bone surface. The Krogh model also predicts dramatic differences in oxygen availability during bone development. Thus, during osteon formation, the modeling equations predict a steep oxygen gradient at the initial stage of development, with the gradient becoming lesser as osteonal layers are added. In contrast, during trabeculum formation, the oxygen gradient is steepest when the diameter of the trabeculum is maximal. Based on these results, it is concluded that significant oxygen gradients exist within cortical and cancellous bone and that the oxygen tension may regulate the physical dimensions of both osteons and bone trabeculae.


Assuntos
Osso e Ossos , Ósteon , Modelos Biológicos , Modelos Teóricos , Oxigênio/metabolismo , Animais , Osso e Ossos/metabolismo , Osso e Ossos/ultraestrutura , Ósteon/metabolismo , Ósteon/ultraestrutura , Humanos
10.
Clin Orthop Relat Res ; 468(8): 2113-21, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20361282

RESUMO

BACKGROUND: Bone grafts are frequently used to supplement bone stock and to establish structural stability. However, graft-associated infection represents a challenging complication leading to increased patient morbidity and healthcare costs. QUESTIONS/PURPOSES: We therefore designed this study to (1) determine if increasing initial S. aureus inoculation of bone allograft results in a proportionate increase in colonization; (2) assess if antibiotics decrease colonization and if antibiotic tethering to allograft alters its ability to prevent bacterial colonization; and (3) determine if covalent modification alters the allograft topography or its biological properties. METHODS: Allograft bone and vancomycin-modified bone (VAN-bone) was challenged with different doses of S. aureus for times out to 24 hours in the presence or absence of solution vancomycin. Bacterial colonization was assessed by fluorescence, scanning electron microscopy (SEM), and by direct colony counting. Cell density and distribution of osteoblast-like cells on control and modified allograft were then compared. RESULTS: Bacterial attachment was apparent within 6 hours with colonization and biofilm formation increasing with time and dose. Solution vancomycin failed to prevent bacterial attachment whereas VAN-bone successfully resisted colonization. The allograft modification did not affect the attachment and distribution of osteoblast-like cells. CONCLUSIONS: Allograft bone was readily colonized by S. aureus and covered by a biofilm with especially florid growth in natural topographic niches. Using a novel covalent modification, allograft bone was able to resist colonization by organisms while retaining the ability to allow adhesion of osteoblastic cells. CLINICAL RELEVANCE: Generation of allograft bone that can resist infection in vivo would be important in addressing one of the most challenging problems associated with the use of allograft, namely infection.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Transplante Ósseo , Próteses e Implantes/microbiologia , Staphylococcus aureus/efeitos dos fármacos , Vancomicina/farmacologia , Antibacterianos/química , Substitutos Ósseos , Técnicas In Vitro , Infecções Relacionadas à Prótese/prevenção & controle , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus/crescimento & desenvolvimento , Transplante Homólogo , Vancomicina/química
11.
Tissue Eng Part A ; 16(6): 2041-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20109017

RESUMO

Infection associated with inert implants is complicated by bacterial biofilm formation that renders the infection antibiotic insensitive. The goal of this investigation was to synthesize and characterize a vancomycin (VAN)-modified bone allograft that could render the tissue inhospitable to bacterial colonization and the establishment of infection. We found that the numbers of primary amines, which could serve as anchors for chemical synthesis, increased with limited demineralization. Using these amines, we coupled two linkers and VAN to bone using Fmoc chemistry. By immunohistochemistry, VAN was abundant on the surface of the allograft; based on elution and measurement of bound antibody, this coupling yielded at least approximately 26 ng VAN/mg bone. The coupled VAN appeared to be permanently bound to the allograft, as it showed no elution in a disk diffusion assay, and, importantly, resisted colonization by Staphylococcus aureus challenges. We suggest that this chimeric construct represents a new generation of antibiotic-modified allografts that provide antibacterial properties.


Assuntos
Antibacterianos/farmacologia , Alicerces Teciduais/química , Transplante Homólogo/métodos , Vancomicina/farmacologia , Aminas/química , Antibacterianos/química , Humanos , Imuno-Histoquímica , Microscopia Confocal , Espectrofotometria Atômica , Staphylococcus aureus/efeitos dos fármacos , Vancomicina/química
12.
Arthritis Rheum ; 60(5): 1406-15, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19404942

RESUMO

OBJECTIVE: We have previously demonstrated that the transcription factor hypoxia-inducible factor 1 (HIF-1) promotes the onset of autophagy in chondrocytes. The overall goal of this study was to test the hypothesis that another HIF family transcription factor, HIF-2, modulates the induction of autophagy by chondrocytes. METHODS: Expression of HIF-1, HIF-2, and light chain 3 (LC3) in human and murine articular cartilage was visualized by immunohistochemistry. Suppression of HIF-2 was achieved using small interfering RNA technology. Assessments of autophagic flux and lysosomal activity, as well as ultrastructural analysis, were performed in chondrocytes in cell culture. RESULTS: HIF-2 was expressed abundantly by cells in human and murine articular cartilage and in the cartilage of mineralizing vertebrae from neonatal mice. Protein levels were reduced in articular cartilage from older mice, in end-plate cartilage from mice, and in chondrocytes from human osteoarthritic (OA) cartilage. HIF-2 was robustly expressed in the prehypertrophic cells of mouse growth cartilage. When HIF-2alpha was silenced, the generation of reactive oxygen species was found to be elevated, with a concomitant decrease in catalase and superoxide dismutase activity. Suppression of HIF-2 was associated with decreased Akt-1 and mammalian target of rapamycin activities, reduced Bcl-xL expression, and a robust autophagic response, even under nutrient-replete conditions. In these silenced chondrocytes, HIF-1 expression was elevated. Decreased HIF-2 expression was associated with autophagy in OA tissues and aging cartilage samples. The autophagic response of chondrocytes in HIF-2alpha-knockout mouse growth plate showed an elevated autophagic response throughout the plate. CONCLUSION: Based on these observations, we conclude that HIF-2 is a potent regulator of autophagy in maturing chondrocytes. Our data suggest that this protein acts as a brake on the autophagy-accelerator function of HIF-1.


Assuntos
Autofagia/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Condrócitos/fisiologia , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/análise , Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Catalase/análise , Células Cultivadas , Condrócitos/ultraestrutura , Humanos , Fator 1 Induzível por Hipóxia/análise , Fator 1 Induzível por Hipóxia/fisiologia , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/análise , Superóxido Dismutase/análise
13.
Plasmid ; 62(1): 50-5, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19389425

RESUMO

Cartilage development and function are dependent on a temporally integrated program of gene expression. With the advent of RNA interference (RNAi), artificial control of these complex programs becomes a possibility, limited only by the ability to regulate and express the RNAi. Using existing methods for production of RNAi's, we have constructed a plasmid-based short hairpin RNA (shRNA) expression system under control of the human pol III H1 promoter and supplemented this promoter with DNA binding sites for the cartilage-specific transcription factor Sox9. The resulting shRNA expression system displays robust, Sox9-dependent gene silencing. Dependence on Sox9 expression was confirmed by electrophoretic mobility shift assays. The ability of the system to regulate heterologously expressed Sox9 was demonstrated by Western blot, as a function of both Sox9 to shRNA ratio, as well as time from transfection. This novel expression system supports auto-regulatory gene silencing, providing a tissue-specific feedback mechanism for temporal control of gene expression. Its applications for both basic mechanistic studies and therapeutic purposes should facilitate the design and implementation of innovative tissue engineering strategies.


Assuntos
Inativação Gênica , Técnicas Genéticas , Conformação de Ácido Nucleico , RNA Interferente Pequeno/genética , Fatores de Transcrição SOX9/metabolismo , Linhagem Celular , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Humanos , Plasmídeos/genética , Regiões Promotoras Genéticas/genética , RNA Interferente Pequeno/química , Fatores de Tempo
14.
Clin Orthop Relat Res ; 467(7): 1678-87, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19365697

RESUMO

Periprosthetic infection is increasingly prevalent in orthopaedics with infection rates of 2% to 15% after total hip arthroplasty. To effectively decrease bacterial attachment, colonization, and subsequent development of periprosthetic infection, we previously described a method to covalently bond vancomycin to smooth Ti alloy surfaces. To attach vancomycin, the Ti surface is first passivated to create a fresh oxide layer. Previously, passivation has been achieved with an H2SO4/H2O2 etch that can destroy the topography of the underlying implant. Passivation by hydrothermal aging as well as by H2SO4/H2O2 incubation produced a robust oxide layer, but only hydrothermal aging left the geometry unaltered. These hydrothermally passivated Kirschner wires and smooth or beaded Ti surfaces were chemically coupled with vancomycin. Antibiotic-coupled samples representing all three geometries were uniformly covered with antibiotic, resisted colonization by Staphylococcus aureus for longer than 8 hours, and retained their biocompatibility as assessed by normal attachment and morphology of preosteocytic MLO-A5 cells. Using this technique, we believe it is possible to passivate many complex implant designs/geometries as a first step toward covalent bonding of antibiotics or other bioactive factors.


Assuntos
Antibacterianos/química , Teste de Materiais , Procedimentos Ortopédicos , Infecções Relacionadas à Prótese/prevenção & controle , Titânio/química , Vancomicina/química , Aderência Bacteriana/efeitos dos fármacos , Temperatura Alta , Humanos , Peróxido de Hidrogênio/química , Interações Hidrofóbicas e Hidrofílicas , Microscopia Eletrônica de Varredura , Oxirredução , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Ácidos Sulfúricos/química
15.
Angew Chem Int Ed Engl ; 48(20): 3677-80, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19353613

RESUMO

As easy as 1, 2, 3: A palladium-catalyzed three-component coupling generates alpha,beta-unsaturated gamma-amino acids in a single step (see scheme). The reaction is believed to involve migration of a vinyl substituent to a highly electrophilic palladium carbene. Unlike previous synthetic approaches, this synthesis provides access to gamma-amino acids with non-natural side chains.

16.
J Orthop Res ; 27(6): 701-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19051247

RESUMO

Peri-prosthetic infection remains a serious complication of joint replacement surgery. Herein, we demonstrate that a vancomycin-containing sol-gel film on Ti alloy rods can successfully treat bacterial infections in an animal model. The vancomycin-containing sol-gel films exhibited predictable release kinetics, while significantly inhibiting S. aureus adhesion. When evaluated in a rat osteomyelitis model, microbiological analysis indicated that the vancomycin-containing sol-gel film caused a profound decrease in S. aureus number. Radiologically, while the control side showed extensive bone degradation, including abscesses and an extensive periosteal reaction, rods coated with the vancomycin-containing sol-gel film resulted in minimal signs of infection. MicroCT analysis confirmed the radiological results, while demonstrating that the vancomycin-containing sol-gel film significantly protected dense bone from resorption and minimized remodeling. These results clearly demonstrate that this novel thin sol-gel technology can be used for the targeted delivery of antibiotics for the treatment of periprosthetic as well as other bone infections.


Assuntos
Antibacterianos/farmacocinética , Materiais Revestidos Biocompatíveis , Osteomielite/tratamento farmacológico , Infecções Relacionadas à Prótese/tratamento farmacológico , Vancomicina/farmacocinética , Animais , Densidade Óssea , Pinos Ortopédicos/microbiologia , Reabsorção Óssea/diagnóstico por imagem , Modelos Animais de Doenças , Fêmur/microbiologia , Fêmur/cirurgia , Géis , Osteogênese , Osteomielite/diagnóstico por imagem , Infecções Relacionadas à Prótese/diagnóstico por imagem , Radiografia , Ratos , Infecções Estafilocócicas/diagnóstico por imagem , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/efeitos dos fármacos , Titânio
17.
Biomaterials ; 29(35): 4684-90, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18814909

RESUMO

Peri-prosthetic infections are notoriously difficult to treat as the biomaterial implant is ideal for bacterial adhesion and biofilm formation, resulting in decreased antibiotic sensitivity. Previously, we reported that vancomycin covalently attached to a Ti alloy surface (Vanc-Ti) could prevent bacterial colonization. Herein we examine the effect of this Vanc-Ti surface on Staphylococci epidermidis, a Gram-positive organism prevalent in orthopaedic infections. By direct colony counting and fluorescent visualization of live bacteria, S. epidermidis colonization was significantly inhibited on Vanc-Ti implants. In contrast, the gram-negative organism Escherichia coli readily colonized the Vanc-Ti rod, suggesting retention of antibiotic specificity. By histochemical and SEM analysis, Vanc-Ti prevented S. epidermidis biofilm formation, even in the presence of serum. Furthermore, when challenged multiple times with S. epidermidis, Vanc-Ti rods resisted bacterial colonization. Finally, when S. epidermidis was continuously cultured in the presence of Vanc-Ti, the bacteria maintained a Vanc sensitivity equivalent to the parent strain. These findings indicate that antibiotic derivatization of implants can result in a surface that can resist bacterial colonization. This technology holds great promise for the prevention and treatment of periprosthetic infections.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Infecções Relacionadas à Prótese/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus epidermidis/efeitos dos fármacos , Staphylococcus epidermidis/crescimento & desenvolvimento , Titânio , Vancomicina/farmacologia , Ligas/química , Animais , Antibacterianos/administração & dosagem , Infecções Relacionadas à Prótese/microbiologia , Infecções Estafilocócicas/microbiologia , Titânio/química , Vancomicina/química
18.
Bone ; 43(1): 25-31, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18485858

RESUMO

Oxygen availability is a critical signal for proper development of many tissues, however there is limited knowledge of its role in the maturation of bone cells. To test the hypothesis that low pO2 regulates bone cell mineralization, MLO-A5 and MLO-Y4 cells were cultured in monolayer and three-dimensional alginate scaffolds in hypoxia (2% O2) or normoxia (20% O2). Hypoxia reduced mineralization and decreased alkaline phosphatase activity of preosteocyte-like MLO-A5 cells in both monolayer and alginate cultures. Similar changes in osteogenic activity were seen when the were subjected to chemical hypoxia. Likewise, Osteocyte-like MLO-Y4 cells also exhibited reduced osteogenic activity in hypoxia relative to normoxic controls. Based on these observations, it is concluded that a low pO2 decreased the mineralization potential of bone cells at both early and late stages of maturation. Since the oxemic state is transduced by the transcription factor, HIF-1alpha, experiments were performed to determine if this protein was responsible for the observed changes in mineral formation. It was noted that when HIF-1alpha was silenced, mineralization activities were not restored. Indeed, in hypoxia, in relationship to wild type controls, the mineralization potential of the knockdown cells was further reduced. Based on these findings, it is concluded that the osteogenic activity of preosteocyte-like cells is dependent on both the O2 tension and the expression of HIF-1alpha.


Assuntos
Calcificação Fisiológica , Osteócitos/citologia , Oxigênio/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Western Blotting , Linhagem Celular , Camundongos , Osteócitos/enzimologia , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Clin Orthop Relat Res ; 462: 200-6, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17572634

RESUMO

Antibiotic concentrations associated with antibiotic bone cements may cause skeletal cell toxicity and prevent fracture healing. We investigated toxicity effects of dose and treatment time after exposure to three antibiotics commonly used in orthopaedic local drug delivery systems. We hypothesized a threshold exists for toxicity of osteoblasts and chondrocytes after treatment with ciprofloxacin, vancomycin, or tobramycin. To test this hypothesis, we first determined whether treatment with antibiotics caused differences in cellular morphology. Cells exposed to ciprofloxacin showed considerable changes in spread, cell membrane, and extensions. We next asked what dosage of antibiotic would cause reductions in osteoblast and chondrocyte cell numbers. Ciprofloxacin at a dose greater than 100 microg/mL and vancomycin and tobramycin at doses greater than 2000 microg/mL severely decreased cellular proliferation. Finally, we questioned whether observed decreases in cell numbers were the result of increased cellular toxicity or senescence. Released lactate dehydrogenase ratios were severely increased in osteoblasts. These data suggest the balance between the targeted microbicidal effects and host cellular toxicity is critical for skeletal cell survival and function.


Assuntos
Antibacterianos/administração & dosagem , Antibacterianos/toxicidade , Condrócitos/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Osteoblastos/efeitos dos fármacos , Células 3T3 , Animais , Materiais Biocompatíveis , Cimentos Ósseos , Contagem de Células , Membrana Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Condrócitos/enzimologia , Condrócitos/patologia , Ciprofloxacina/toxicidade , Relação Dose-Resposta a Droga , L-Lactato Desidrogenase/metabolismo , Camundongos , Osteoblastos/enzimologia , Osteoblastos/patologia , Tobramicina/toxicidade , Vancomicina/toxicidade
20.
Clin Orthop Relat Res ; 461: 81-7, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17549031

RESUMO

Despite improved strategies for treating periprosthetic infection, current antibiotic delivery approaches are imperfect and can result in bacterial resistance and recalcitrant bio-films. To address the issues, we developed a covalently linked vancomycin-titanium implant interface that prevents and possibly eliminates bacterial colonization. We determined the amount of vancomycin immobilized on the titanium surface and assessed vancomycin stability and activity over time. When incubated with Staphylococcus aureus, the vancomycin-titanium surface showed an almost complete absence of adherent bacteria. To determine if continual exposure to vancomycin-titanium would cause decreased susceptibility to the antibiotic, S. aureus was incubated with vancomycin-titanium for 1 week or 4 weeks; these bacteria did not show an increased minimum inhibitory concentration for vancomycin. We tested the long-term stability of the vancomycin-titanium surface by incubation in phosphate-buffered saline for 11 months and then challenging the surface with S. aureus. Fluorescent staining for bacteria indicated the vancomycin-titanium retained its bactericidal activity. Finally, osteoblasts seeded on the vancomycin-titanium surface exhibited no change in viability, indicating the surface supports bone cell adhesion. Based on these observations, covalent modification of the titanium surface with an antibiotic may be viewed as a potential new tool in preventing or eliminating periprosthetic infection.


Assuntos
Antibacterianos/administração & dosagem , Prótese de Quadril/microbiologia , Infecções Relacionadas à Prótese/prevenção & controle , Titânio/química , Vancomicina/administração & dosagem , Adesão Celular , Prótese de Quadril/efeitos adversos , Nanotubos , Desenho de Prótese , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...