Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Sci Rep ; 12(1): 18387, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36319854

RESUMEN

Currently, implants are utilized clinically for bone transplant procedures. However, if infectious osteomyelitis occurs at implant sites, removal of bacteria can be challenging. Moreover, altered blood flow at peri-implant infectious sites can create an anaerobic environment, making it more difficult to treat infection with antibiotics. Thus, it would be beneficial if implants could be modified to exhibit antibacterial activity, even in anaerobic conditions. Here, we show antibacterial activity of silver ions coated on titanium rods, even against the anaerobic bacteria Porphyromonas gingivalis (P. gingivalis), both in vitro and in vivo. Specifically, we implanted silver-coated or control uncoated titanium rods along with P. gingivalis in mouse femoral bone BM cavities and observed significantly inhibited P. gingivalis infection with silver-coated compared with non-coated rods, based on in vivo bio-imaging. Osteonecrosis by infectious osteomyelitis and elevation of the inflammatory factors C-reactive protein and IL-6 promoted by P. gingivalis s were also significantly reduced in the presence of silver-coated rods. Overall, our study indicates that silver ion coating of an implant represents a therapeutic option to prevent associated infection, even in anaerobic conditions or against anaerobic bacteria.


Asunto(s)
Antibacterianos , Bacterias Anaerobias , Materiales Biocompatibles Revestidos , Implantes Experimentales , Osteomielitis , Plata , Animales , Ratones , Antibacterianos/farmacología , Bacterias Anaerobias/efectos de los fármacos , Materiales Biocompatibles Revestidos/farmacología , Iones/farmacología , Osteomielitis/microbiología , Osteomielitis/prevención & control , Plata/farmacología , Titanio/química , Porphyromonas gingivalis/efectos de los fármacos , Implantes Experimentales/efectos adversos , Implantes Experimentales/microbiología , Fémur , Proteína C-Reactiva
2.
Molecules ; 26(14)2021 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-34299652

RESUMEN

Implantable medical devices (IMDs) are susceptible to microbial adhesion and biofilm formation, which lead to several clinical complications, including the occurrence of implant-associated infections. Polylactic acid (PLA) and its composites are currently used for the construction of IMDs. In addition, chitosan (CS) is a natural polymer that has been widely used in the medical field due to its antimicrobial and antibiofilm properties, which can be dependent on molecular weight (Mw). The present study aims to evaluate the performance of CS-based surfaces of different Mw to inhibit bacterial biofilm formation. For this purpose, CS-based surfaces were produced by dip-coating and the presence of CS and its derivatives onto PLA films, as well surface homogeneity were confirmed by contact angle measurements, Fourier transform infrared spectroscopy (FTIR) and scanning electron microscopy (SEM). The antimicrobial activity of the functionalized surfaces was evaluated against single- and dual-species biofilms of Staphylococcus aureus and Pseudomonas aeruginosa. Chitosan-based surfaces were able to inhibit the development of single- and dual-species biofilms by reducing the number of total, viable, culturable, and viable but nonculturable cells up to 79%, 90%, 81%, and 96%, respectively, being their activity dependent on chitosan Mw. The effect of CS-based surfaces on the inhibition of biofilm formation was corroborated by biofilm structure analysis using confocal laser scanning microscopy (CLSM), which revealed a decrease in the biovolume and thickness of the biofilm formed on CS-based surfaces compared to PLA. Overall, these results support the potential of low Mw CS for coating polymeric devices such as IMDs where the two bacteria tested are common colonizers and reduce their biofilm formation.


Asunto(s)
Biopelículas/efectos de los fármacos , Quitosano , Implantes Experimentales/microbiología , Pseudomonas aeruginosa/fisiología , Staphylococcus aureus/fisiología , Biopelículas/crecimiento & desarrollo , Quitosano/química , Quitosano/farmacología , Propiedades de Superficie
3.
J Biomed Mater Res A ; 109(5): 637-648, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32615012

RESUMEN

A prosthetic scaffold development using fluorescent nanofiber is reported for an enhanced reepithelialization in wistar albino rats. In this study, a novel approach was followed to construct the biocompatible fluorescent nanofiber that will be helpful to monitor the tissue regeneration process. Here, a multifunctional carbon quantum dots (CQDs)-embedded electrospun polyacrylonitrile (PAN) nanofiber was fabricated and characterized using standard laboratory techniques. The biodegradation ability was assessed by simulated body fluid thereby analyzing porosity and water absorption capacity of the material. The fluorescent scaffold was tested for cytotoxicity and antimicrobial activity using bacterial and fibroblast cells and fluorescent stability was analyzed by bioimaging of animal and bacterial cells. Tissue regeneration capability of the developed scaffold was evaluated using wistar albino rats. Unlike biomicking scaffolds, the CQDs-embedded PAN-based substrate has given dual support by enhancing reepithelialization without growth factors and acted as an antimicrobial agent to provide contamination free tissue regeneration. Scaffolds were examined by using histostaining techniques and scanning electron microscopy to observe the reepithelialization in the regenerated tissues. The novel approach for developing infection free soft tissue regeneration was found to be phenomenal in scaffold development.


Asunto(s)
Materiales Biocompatibles , Carbono , Regeneración Tisular Dirigida , Puntos Cuánticos/uso terapéutico , Repitelización/efectos de los fármacos , Andamios del Tejido , Resinas Acrílicas , Animales , Materiales Biocompatibles/efectos adversos , Adhesión Celular , Línea Celular , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Fibroblastos/efectos de los fármacos , Implantes Experimentales/efectos adversos , Implantes Experimentales/microbiología , Ensayo de Materiales , Ratones , Microscopía Electrónica de Rastreo , Nanofibras , Puntos Cuánticos/administración & dosificación , Ratas , Ratas Wistar , Piel/lesiones , Propiedades de Superficie , Andamios del Tejido/efectos adversos , Humectabilidad
4.
Mater Sci Eng C Mater Biol Appl ; 109: 110573, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32228937

RESUMEN

Implant associated infections can result in devastating consequences for patients. One major cause is the formation of bacterial biofilms, which result in increased resistance against antimicrobial therapeutics. A reduction of implant associated infections can be achieved by functionalization of implant surfaces. The generation of three dimensional surface structures by femtosecond laser ablation is one method to fabricate bacterial repellent large scaled surfaces without altering the material chemical composition. The challenge is to reduce bacterial growth while improving cellular ongrowth. For this purpose, spike structures were created as small as possible by used fabrication method on cubic Ti90/Al6/V4-rods and their effectiveness against bacterial colonization was compared to unstructured Ti90/Al6/V4-rods. Rods were implanted in the rat tibia and infected intraoperatively with 103 CFU of Staphylococcus aureus. Besides clinical behaviour and lameness, the vital bacterial biomass, morphological appearance and the volume of eukaryotic cells were determined on the implant surface after 21 days. Bone alterations were examined by radiological and histological techniques. Unexpectedly, the laser-structured implants did not show a lower bacterial load on the implant surface and less severe infection related bone and tissue alterations compared to the group without structuring. Simultaneously, a better bony integration and a higher cellular colonization with eukaryotic cells was detected on the laser-structured implants. These findings don't support the previous in vitro results. Nevertheless, the strong integration into the bone is a powerful argument for further surface modifications focussing on the improvement of the antibacterial effect. Additionally, our results underline the need for in vivo testing of new materials prior to clinical use.


Asunto(s)
Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Implantes Experimentales/microbiología , Infecciones Relacionadas con Prótesis , Infecciones Estafilocócicas , Staphylococcus aureus/fisiología , Animales , Adhesión Bacteriana/efectos de los fármacos , Rayos Láser , Masculino , Infecciones Relacionadas con Prótesis/tratamiento farmacológico , Infecciones Relacionadas con Prótesis/metabolismo , Infecciones Relacionadas con Prótesis/patología , Ratas , Ratas Endogámicas Lew , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/patología , Uracilo/análogos & derivados
5.
PLoS Pathog ; 16(3): e1008354, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32142554

RESUMEN

Biofilm-associated prosthetic joint infections (PJIs) cause significant morbidity due to their recalcitrance to immune-mediated clearance and antibiotics, with Staphylococcus aureus (S. aureus) among the most prevalent pathogens. We previously demonstrated that S. aureus biofilm-associated monocytes are polarized to an anti-inflammatory phenotype and the adoptive transfer of pro-inflammatory macrophages attenuated biofilm burden, highlighting the critical role of monocyte/macrophage inflammatory status in dictating biofilm persistence. The inflammatory properties of leukocytes are linked to their metabolic state, and here we demonstrate that biofilm-associated monocytes exhibit a metabolic bias favoring oxidative phosphorylation (OxPhos) and less aerobic glycolysis to facilitate their anti-inflammatory activity and biofilm persistence. To shift monocyte metabolism in vivo and reprogram cells to a pro-inflammatory state, a nanoparticle approach was utilized to deliver the OxPhos inhibitor oligomycin to monocytes. Using a mouse model of S. aureus PJI, oligomycin nanoparticles were preferentially internalized by monocytes, which significantly reduced S. aureus biofilm burden by altering metabolism and promoting the pro-inflammatory properties of infiltrating monocytes as revealed by metabolomics and RT-qPCR, respectively. Injection of oligomycin alone had no effect on monocyte metabolism or biofilm burden, establishing that intracellular delivery of oligomycin is required to reprogram monocyte metabolic activity and that oligomycin lacks antibacterial activity against S. aureus biofilms. Remarkably, monocyte metabolic reprogramming with oligomycin nanoparticles was effective at clearing established biofilms in combination with systemic antibiotics. These findings suggest that metabolic reprogramming of biofilm-associated monocytes may represent a novel therapeutic approach for PJI.


Asunto(s)
Biopelículas/efectos de los fármacos , Reprogramación Celular/efectos de los fármacos , Implantes Experimentales/microbiología , Monocitos/metabolismo , Oligomicinas/farmacología , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/fisiología , Animales , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Ratones , Monocitos/patología , Fosforilación Oxidativa/efectos de los fármacos , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/patología
6.
ACS Appl Mater Interfaces ; 11(29): 25691-25701, 2019 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-31264401

RESUMEN

The greatest bottleneck for photothermal antibacterial therapy could be the difficulty in heating the infection site directly and specifically to evade the unwanted damage for surrounding healthy tissues. In recent years, infectious microenvironments (IMEs) have been increasingly recognized as a crucial contributor to bacterial infections. Here, based on the unique IMEs and rhenium trioxide (ReO3) nanocubes (NCs), a new specific photothermal antibacterial strategy is reported. These NCs synthesized by a rapid and straightforward space-confined on-substrate approach have good biocompatibility and exhibit efficient photothermal antibacterial ability. Especially when they are utilized in antibiofilm, the expression levels of biofilm-related genes (icaA, fnbA, atlE, and sarA for Staphylococcus aureus) can be effectively inhibited to block bacterial adhesion and formation of biofilm. Importantly, the ReO3 NCs can transform into hydrogen rhenium bronze (HxReO3) in an aqueous environment, making them relatively stable within the low pH of IMEs for photothermal therapy, while rapidly degradable within the surrounding healthy tissues to decrease photothermal damage. Note that under phosphate-buffered saline (PBS) at pH 7.4 without assistant conditions, these ReO3 NCs have the highest degradation rate among all known degradable inorganic photothermal nanoagents. This special and IME-sensitive selective degradability of the ReO3 NCs not only facilitates safe, efficient, and specific elimination of implant-related infections, but also enables effective body clearance after therapy. Solely containing the element (Re) whose atomic number is higher than clinic-applied iodine in all reported degradable inorganic photothermal nanoagents under the PBS (pH 7.4) without any assistant condition, the ReO3 NCs with high X-ray attenuation ability could be further applied to X-ray computed tomography imaging-guided therapy against implant-related infections. The present work described here is the first to adopt degradable inorganic photothermal nanoagents to achieve specific antibacterial therapy and inspires other therapies on this concept.


Asunto(s)
Antibacterianos , Hipertermia Inducida , Implantes Experimentales/microbiología , Nanoestructuras/química , Fototerapia , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus/fisiología , Animales , Antibacterianos/química , Antibacterianos/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Óxidos/química , Renio/química
7.
ACS Appl Mater Interfaces ; 11(27): 24588-24597, 2019 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-31199619

RESUMEN

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.


Asunto(s)
Antibacterianos , Materiales Biocompatibles Revestidos , Fibroblastos , Implantes Experimentales/microbiología , Nanodiamantes/química , Osteoblastos , Staphylococcus aureus/crecimiento & desarrollo , Titanio/química , Antibacterianos/química , Antibacterianos/farmacología , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Fibroblastos/metabolismo , Fibroblastos/microbiología , Fibroblastos/patología , Humanos , Osteoblastos/metabolismo , Osteoblastos/microbiología , Osteoblastos/patología
8.
J Biomed Mater Res B Appl Biomater ; 107(1): 169-177, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29573163

RESUMEN

Periprosthetic infection via skin-implant interface is a leading cause of failures and revisions in direct skeletal attachment of limb prostheses. Implants with deep porosity fabricated with skin and bone integrated pylons (SBIP) technology allow for skin ingrowth through the implant's structure creating natural barrier against infection. However, until the skin cells remodel in all pores of the implant, additional care is required to prevent from entering bacteria to the still nonoccupied pores. Temporary silver coating was evaluated in this work as a means to provide protection from infection immediately after implantation followed by dissolution of silver layer in few weeks. A sputtering coating with 1 µm thickness was selected to be sufficient for fighting infection until the deep ingrowth of skin in the porous structure of the pylon is completed. In vitro study showed less bacterial (Staphylococcus aureus, Staphylococcus epidermidis, and Pseudomonas aeruginosa) growth on silver coated tablets compared to the control group. Analysis of cellular density of MG-63 cells, fibroblasts, and mesenchymal stem cells (MSCs) showed that silver coating did not inhibit the cell growth on the implants and did not affect cellular functional activity. The in vivo study did not show any postoperative complications during the 6-month observation period in the model of above-knee amputation in rabbits when SBIP implants, either silver-coated or untreated were inserted into the bone residuum. Three-phase scintigraphy demonstrated angiogenesis in the pores of the pylons. The findings suggest that a silver coating with well-chosen specifications can increase the safety of porous implants for direct skeletal attachment. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 107B: 169-177, 2019.


Asunto(s)
Antibacterianos/química , Bacterias/crecimiento & desarrollo , Infecciones Bacterianas , Interfase Hueso-Implante , Materiales Biocompatibles Revestidos/química , Implantes Experimentales/microbiología , Plata/química , Piel , Animales , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/patología , Interfase Hueso-Implante/microbiología , Interfase Hueso-Implante/patología , Línea Celular Tumoral , Humanos , Masculino , Porosidad , Conejos , Piel/microbiología , Piel/patología
9.
J Biomed Mater Res B Appl Biomater ; 107(4): 1095-1106, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30332531

RESUMEN

The development of an infection is a major complication for some patients with implanted biomaterials. Whether the material or surface composition of the used biomaterial influences infection has not been directly compared for key biomaterials currently in use in human patients. We conducted a thorough in vitro and in vivo investigation using titanium (Ti) and polyether-ether-ketone (PEEK) as both commercially available and as modified equivalents (surface polished Ti, and oxygen plasma treated PEEK). Complement activation and cytokine secretion of cell of the immune system was assessed in vitro for all materials in the absence and presence of bacterial stimulants. In a follow-up in vivo study, we monitored bacterial infection associated with clinically available and standard Ti and PEEK inoculated with Staphylococcus aureus. Complement activation was affected by material choice in the absence of bacterial stimulation, although the material based differences were largely lost upon bacterial stimulation. In the in vivo study, the bacterial burden, histological response and cytokine secretion suggests that there is no significant difference between both PEEK and Ti. In conclusion, the underlying material has a certain impact in the absence of bacterial stimulation, however, in the presence of bacterial stimulation, bacteria seem to dictate the responses in a manner that overshadows the influence of material surface properties. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1095-1106, 2019.


Asunto(s)
Enfermedades Óseas Infecciosas , Implantes Experimentales/microbiología , Cetonas/química , Ensayo de Materiales , Polietilenglicoles/química , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Animales , Benzofenonas , Enfermedades Óseas Infecciosas/inmunología , Enfermedades Óseas Infecciosas/microbiología , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Oseointegración , Polímeros , Infecciones Estafilocócicas/patología
10.
Acta Biomater ; 73: 250-262, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29679779

RESUMEN

In this work, we define the requirements for a human cell-based osteomyelitis model which overcomes the limitations of state of the art animal models. Osteomyelitis is a severe and difficult to treat infection of the bone that develops rapidly, making it difficult to study in humans. We have developed a 3D in vitro model of the bone marrow, comprising a macroporous material, human hematopoietic stem and progenitor cells (HSPCs) and mesenchymal stromal cells (MSCs). Inclusion of biofilms grown on an implant into the model system allowed us to study the effects of postoperative osteomyelitis-inducing bacteria on the bone marrow. The bacteria influenced the myeloid differentiation of HSPCs as well as MSC cytokine expression and the MSC ability to support HSPC maintenance. In conclusion, we provide a new 3D in vitro model which meets all the requirements for investigating the impact of osteomyelitis. STATEMENT OF SIGNIFICANCE: Implant-associated osteomyelitis is a persistent bacterial infection of the bone which occurs in many implant patients and can result in functional impairments or even entire loss of the extremity. Nevertheless, surprisingly little is known on the triangle interaction between implant material, bacterial biofilm and affected bone tissue. Closing this gap of knowledge would be crucial for the fundamental understanding of the disease and the development of novel treatment strategies. For this purpose, we developed the first biomaterial-based system that is able to mimic implant-associated osteomyelitis outside of the body, thus, opening the avenue to study this fatal disease in the laboratory.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Materiales Biomiméticos/farmacología , Enfermedades de la Médula Ósea , Hematopoyesis , Implantes Experimentales/microbiología , Staphylococcus aureus Resistente a Meticilina/fisiología , Modelos Biológicos , Osteomielitis , Infecciones Estafilocócicas , Enfermedades de la Médula Ósea/metabolismo , Enfermedades de la Médula Ósea/microbiología , Enfermedades de la Médula Ósea/patología , Células Cultivadas , Humanos , Implantes Experimentales/efectos adversos , Osteomielitis/metabolismo , Osteomielitis/microbiología , Osteomielitis/patología , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología
11.
J Biomed Mater Res B Appl Biomater ; 106(1): 221-227, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28130825

RESUMEN

Magnesium alloys hold great promise for developing orthopedic implants that are biocompatible, biodegradable, and mechanically similar to bone tissue. This study evaluated the in vitro and in vivo antimicrobial properties of magnesium-9%aluminum-1%zinc (AZ91) and commercially pure titanium (cpTi) against Acinetobacter baumannii (Ab307). The in vitro results showed that as compared to cpTi, incubation with AZ91 significantly reduced both the planktonic (cpTi = 3.45e8, AZ91 = 8.97e7, p < 0.001) colony forming units (CFU) and biofilm-associated (cpTi = 3.89e8, AZ91 = 1.78e7, p = 0.01) CFU of Ab307. However, in vivo results showed no significant differences in the CFU enumerated from the cpTi and AZ91 implants following a 1-week implantation in an established rodent model of Ab307 implant associated infection (cpTi = 5.23e3, AZ91 = 2.46e3, p = 0.29). It is proposed that the in vitro results were associated with an increased pH in the bacterial culture as a result of the AZ91 corrosion process. The robust in vivo buffering capacity likely diminished this corrosion associated pH antimicrobial effect. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 221-227, 2018.


Asunto(s)
Acinetobacter baumannii/crecimiento & desarrollo , Aleaciones/farmacología , Antiinfecciosos/farmacología , Implantes Experimentales/microbiología , Magnesio/farmacología , Aleaciones/química , Animales , Antiinfecciosos/química , Ratas , Ratas Long-Evans
12.
Acta Biomater ; 58: 550-560, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28571692

RESUMEN

Prevention of orthopedic device related infection (ODRI) using antibiotics has met with limited amount of success and is still a big concern during post-surgery. As an alternative, use of silver as an antibiotic treatment to prevent surgical infections is being used due to the well-established antimicrobial properties of silver. However, in most cases silver is used in particulate form with wound dressings or with short-term devices such as catheters but not with load-bearing implants. We hypothesize that strongly adherent silver to load-bearing implants can offer longer term solution to infection in vivo. Keeping that in mind, the focus of this study was to understand the long term release study of silver ions for a period of minimum 6months from silver coated surface modified porous titanium implants. Implants were fabricated using a LENS™ system, a powder based additive manufacturing technique, with at least 25% volume porosity, with and without TiO2 nanotubes in phosphate buffer saline (pH 7.4) to see if the total release of silver ions is within the toxic limit for human cells. Considering the fact that infection sites may reduce the local pH, silver release was also studied in acetate buffer (pH 5.0) for a period of 4weeks. Along with that, the osseointegrative properties as well as cytotoxicity of porous titanium implants were assessed in vivo for a period of 12weeks using a rat distal femur model. In vivo results indicate that porous titanium implants with silver coating show comparable, if not better, biocompatibility and bonding at the bone-implant interface negating any concerns related to toxicity related to silver to normal cells. The current research is based on our recently patented technology, however focused on understanding longer-term silver release to mitigate infection related problems in load-bearing implants that can even arise several months after the surgery. STATEMENT OF SIGNIFICANCE: Prevention of orthopedic device related infection using antibiotics has met with limited success and is still a big concern during post-surgery. Use of silver as an antibiotic treatment to prevent surgical infections is being explored due to the well-established antimicrobial properties of silver. However, in most cases silver is used in particulate form with wound dressings or with short-term devices such as catheters but not with load-bearing implants. We hypothesize that strongly adherent silver to load-bearing implants can offer longer-term solution towards infection in vivo. Keeping that in mind, the focus of this study was to understand the long-term release of silver ions, for a period of minimum 6months, from silver coated surface modified porous titanium implants.


Asunto(s)
Materiales Biocompatibles Revestidos , Fémur/cirugía , Implantes Experimentales/microbiología , Plata , Infección de la Herida Quirúrgica/prevención & control , Titanio/química , Animales , Sustitutos de Huesos/química , Sustitutos de Huesos/farmacocinética , Materiales Biocompatibles Revestidos/farmacocinética , Masculino , Porosidad , Ratas , Ratas Sprague-Dawley , Plata/química , Plata/farmacocinética
13.
Proc Natl Acad Sci U S A ; 114(26): E5094-E5102, 2017 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-28607050

RESUMEN

Infection is a major complication of implantable medical devices, which provide a scaffold for biofilm formation, thereby reducing susceptibility to antibiotics and complicating treatment. Hematogenous implant-related infections following bacteremia are particularly problematic because they can occur at any time in a previously stable implant. Herein, we developed a model of hematogenous infection in which an orthopedic titanium implant was surgically placed in the legs of mice followed 3 wk later by an i.v. exposure to Staphylococcus aureus This procedure resulted in a marked propensity for a hematogenous implant-related infection comprised of septic arthritis, osteomyelitis, and biofilm formation on the implants in the surgical legs compared with sham-operated surgical legs without implant placement and with contralateral nonoperated normal legs. Neutralizing human monoclonal antibodies against α-toxin (AT) and clumping factor A (ClfA), especially in combination, inhibited biofilm formation in vitro and the hematogenous implant-related infection in vivo. Our findings suggest that AT and ClfA are pathogenic factors that could be therapeutically targeted against Saureus hematogenous implant-related infections.


Asunto(s)
Anticuerpos Antibacterianos/farmacología , Anticuerpos Neutralizantes/farmacología , Artritis Infecciosa , Biopelículas/efectos de los fármacos , Implantes Experimentales/microbiología , Osteomielitis , Infecciones Estafilocócicas , Staphylococcus aureus/fisiología , Animales , Artritis Infecciosa/tratamiento farmacológico , Artritis Infecciosa/etiología , Artritis Infecciosa/microbiología , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Osteomielitis/tratamiento farmacológico , Osteomielitis/etiología , Osteomielitis/microbiología , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/etiología , Infecciones Estafilocócicas/microbiología , Titanio
14.
Adv Healthc Mater ; 6(11)2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28321991

RESUMEN

Joint replacement surgery is associated with significant morbidity and mortality following infection with either methicillin-resistant Staphylococcus aureus (MRSA) or Staphylococcus epidermidis. These organisms have strong biofilm-forming capability in deep wounds and on prosthetic surfaces, with 103 -104 microbes resulting in clinically significant infections. To inhibit biofilm formation, we developed 3D titanium structures using selective laser melting and then coated them with a silver nanolayer using atomic layer deposition. On bare titanium scaffolds, S. epidermidis growth was slow but on silver-coated implants there were significant further reductions in both bacterial recovery (p < 0.0001) and biofilm formation (p < 0.001). MRSA growth was similarly slow on bare titanium scaffolds and not further affected by silver coating. Ultrastructural examination and viability assays using either human bone or endothelial cells, demonstrated strong adherence and growth on titanium-only or silver-coated implants. Histological, X-ray computed microtomographic, and ultrastructural analyses revealed that silver-coated titanium scaffolds implanted into 2.5 mm defects in rat tibia promoted robust vascularization and conspicuous bone ingrowth. We conclude that nanolayer silver of titanium implants significantly reduces pathogenic biofilm formation in vitro, facilitates vascularization and osseointegration in vivo making this a promising technique for clinical orthopedic applications.


Asunto(s)
Sustitutos de Huesos/química , Materiales Biocompatibles Revestidos/química , Implantes Experimentales/microbiología , Staphylococcus aureus Resistente a Meticilina/crecimiento & desarrollo , Nanoestructuras/química , Neovascularización Fisiológica , Plata/química , Staphylococcus epidermidis/crecimiento & desarrollo , Titanio/química , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratas , Ratas Wistar , Tibia/lesiones , Tibia/metabolismo , Tibia/microbiología , Tibia/patología
15.
J Biomed Mater Res A ; 105(1): 62-72, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27513443

RESUMEN

Implantation of a biomaterial provides an adhesion substratum both to host cell integration and to contaminating bacteria. We studied simultaneous competitive adhesion of Staphylococcus aureus in serial 1:10 dilutions of 108 colony forming units (CFU)/mL and human osteogenic sarcoma (SaOS-2) or primary osteoblast (hOB) cells, both 1x105 cells/mL, to the surfaces of titanium, polydimethylsiloxane and polystyrene. The bacterial adherence and human cell proliferation, cytotoxicity and production of reactive oxygen species (ROS) were studied using fluorometric (fluorescent microscopy and flow cytometry) and colorimetric methods (MTT, LDH and crystal violet). The bacterial cell viability was also evaluated using the drop plate method. The presence of bacteria resulted in reduced adherence of human cells to the surface of the biomaterials, increased production of ROS, and into increased apoptosis. On the other hand, the presence of either type of human cells was associated with a reduction of bacterial colonization of the biomaterial with Staphylococcus aureus. These results suggest that increasing colonization of the biomaterial surface in vitro by one negatively affects colonization by the other. Host cell integration to an implant surface reduces bacterial contamination, which opens novel opportunities for the design of infection-resistant biomaterials in current implantology and future regenerative medicine. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 62-72, 2017.


Asunto(s)
Dimetilpolisiloxanos/química , Implantes Experimentales/microbiología , Poliestirenos/química , Staphylococcus aureus/crecimiento & desarrollo , Línea Celular Tumoral , Humanos
16.
Mater Sci Eng C Mater Biol Appl ; 68: 701-715, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27524071

RESUMEN

Periprosthetic infection is a consequence of implant insertion procedures and strategies for its prevention involve either an increase in the rate of new bone formation or the release of antibiotics such as vancomycin. In this work we combined both strategies and developed a novel, multifunctional three-dimensional porous scaffold that was produced using hydroxyapatite (HA) and ß-tricalcium phosphate (ß-TCP), coupled with a pectin (PEC)-chitosan (CHIT) polyelectrolyte (PEI), and loaded with vancomycin (VCA). By this approach, a controlled vancomycin release was achieved and serial bacterial dilution test demonstrated that, after 1week, the engineered construct still inhibits the bacterial growth. Degradation tests show an excellent behavior in a physiological and acidic environment (<10% of mass loss). Furthermore, the PEI coating shows an anti-inflammatory response, and good cell proliferation and migration were demonstrated in vitro using osteoblast SAOS-2 cell line. This new engineered construct exhibits excellent properties both as an antibacterial material and as a stimulator of bone formation, which makes it a good candidate to contrast periprosthetic infection.


Asunto(s)
Implantes Experimentales/microbiología , Osteoblastos/microbiología , Infecciones Estafilocócicas/prevención & control , Staphylococcus epidermidis/crecimiento & desarrollo , Andamios del Tejido/química , Vancomicina/química , Animales , Fosfatos de Calcio/química , Línea Celular , Quitosano/química , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacología , Durapatita/química , Ratones , Osteoblastos/metabolismo , Pectinas/química , Porosidad , Vancomicina/farmacología
17.
Rev. Esp. Cir. Ortop. Traumatol. (Ed. Impr.) ; 60(4): 260-266, jul.-ago. 2016. ilus, tab, graf
Artículo en Español | IBECS | ID: ibc-153784

RESUMEN

Objetivo. Evaluar in vivo la actividad bactericida antiestafilocócica del farnesol sobre superficies de Ti6Al4V. Material y métodos. Se desarrolló un modelo experimental de infecciones en biomateriales inoculando Staphylococcus aureus ATCC 29213 en los fémures de 15 ratas wistar. Seguidamente se insertó una aguja de Ti6Al4V impregnada con farnesol 30 mM en el fémur estudio y una aguja control en el fémur control. Para valorar la eficacia bactericida se compararon las medianas de unidades formadoras de colonias recuperadas después de la inoculación en el grupo estudio y en el grupo control, para diferentes tiempos de eutanasia y tamaño de inóculos. Resultados. La mediana expresada en Log10 de los recuentos de UFC obtenidos en agujas de titanio con farnesol fue de 4,26 y en agujas sin farnesol, controles, fue de 4,86. Esta diferencia, al aplicar la prueba de t de Student para muestras relacionadas, resultó ser estadísticamente significativa (p = 0,001). La reducción mediana obtenida en las agujas con farnesol respecto a las agujas control fue del 74%. Conclusiones. El tratamiento con farnesol de agujas de Ti6Al4V, a una concentración de 30 mM, parece disminuir la tasa de colonización por Staphylococcus aureus en dichas agujas (AU)


Objective. To evaluate the in vivo anti-staphylococcal bactericidal activity of farnesol on Ti6Al4V surfaces. Material and methods. An experimental model of infection in biomaterials was developed by inoculation of Staphylococcus aureus ATCC 29213 into the canal of both femurs of 15 Wistar rats. A Ti6Al4V pin impregnated with 30 mM of farnesol was inserted into study femur, and a Ti6Al4V control was inserted into the control femur. To evaluate the bactericidal efficacy, a comparison was made between the median of the colony forming units recovered after inoculation in the study group and the control group for different times of euthanasia and inoculum size. Results. The median expressed as Log10 CFU counts obtained with farnesol titanium pin was 4.26, and in control group, it was 4.86, which was statistically significant (P=.001) on applying the Student t test for related samples. The median reduction obtained in farnesol pins relative to the control was 74%. Conclusions. Treatment with farnesol 30 mM on Ti6Al4V pins appears to decrease the rate of colonisation by Staphylococcus aureus (AU)


Asunto(s)
Animales , Masculino , Femenino , Ratas , Implantes Experimentales/microbiología , Implantes Experimentales , Modelos Animales , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/uso terapéutico , Agujas , Actividad Bactericida de la Sangre , Staphylococcus aureus , Staphylococcus aureus/aislamiento & purificación , Fijación Interna de Fracturas/tendencias , Fijación Interna de Fracturas/veterinaria
18.
Lab Anim (NY) ; 45(5): 180-6, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27096188

RESUMEN

The advent of cranial implants revolutionized primate neurophysiological research because they allow researchers to stably record neural activity from monkeys during active behavior. Cranial implants have improved over the years since their introduction, but chronic implants still increase the risk for medical complications including bacterial contamination and resultant infection, chronic inflammation, bone and tissue loss and complications related to the use of dental acrylic. These complications can lead to implant failure and early termination of study protocols. In an effort to reduce complications, we describe several refinements that have helped us improve cranial implants and the wellbeing of implanted primates.


Asunto(s)
Implantes Experimentales/efectos adversos , Macaca mulatta/cirugía , Cráneo/cirugía , Resinas Acrílicas/efectos adversos , Animales , Craneotomía/efectos adversos , Cementos Dentales/efectos adversos , Implantes Experimentales/microbiología , Imagen por Resonancia Magnética , Enfermedades de los Monos/microbiología , Enfermedades de los Monos/prevención & control , Neurofisiología/instrumentación , Neurofisiología/métodos , Complicaciones Posoperatorias/veterinaria , Infección de la Herida Quirúrgica/microbiología , Infección de la Herida Quirúrgica/prevención & control , Infección de la Herida Quirúrgica/veterinaria , Cicatrización de Heridas
19.
Clin Vaccine Immunol ; 22(5): 467-76, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25694647

RESUMEN

Propionibacterium acnes is well-known as a human skin commensal but can also act as an invasive pathogen causing implant-associated infections. In order to resolve these types of P. acnes infections, the implants must be removed, due to the presence of an established biofilm that is recalcitrant to antibiotic therapy. In order to identify those P. acnes proteins produced in vivo during a biofilm infection, we established a rabbit model of implant-associated infection with this pathogen. P. acnes biofilms were anaerobically grown on dextran beads that were then inoculated into the left tibias of rabbits. At 4 weeks postinoculation, P. acnes infection was confirmed by radiograph, histology, culture, and PCR. In vivo-produced and immunogenic P. acnes proteins were detected on Western blot using serum samples from rabbits infected with P. acnes after these bacterial proteins were separated by two-dimensional gel electrophoresis. Those proteins that bound host antibodies were then isolated and identified by tandem mass spectrometry. Radiographs and histology demonstrated a disruption in the normal bone architecture and adherent biofilm communities in those animals with confirmed infections. A total of 24 immunogenic proteins were identified; 13 of these proteins were upregulated in both planktonic and biofilm modes, including an ABC transporter protein. We successfully adapted a rabbit model of implant-associated infection for P. acnes to identify P. acnes proteins produced during a chronic biofilm-mediated infection. Further studies are needed to evaluate the potential of these proteins for either a diagnostic test or a vaccine to prevent biofilm infections caused by P. acnes.


Asunto(s)
Proteínas Bacterianas/aislamiento & purificación , Biopelículas , Infecciones por Bacterias Grampositivas/microbiología , Propionibacterium acnes/química , Propionibacterium acnes/inmunología , Infecciones Relacionadas con Prótesis/microbiología , Proteómica , Transportadoras de Casetes de Unión a ATP/aislamiento & purificación , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Proteínas Bacterianas/análisis , Proteínas Bacterianas/inmunología , Biopelículas/crecimiento & desarrollo , Modelos Animales de Enfermedad , Electroforesis en Gel Bidimensional , Femenino , Implantes Experimentales/microbiología , Osteomielitis/diagnóstico por imagen , Osteomielitis/microbiología , Propionibacterium acnes/genética , Propionibacterium acnes/crecimiento & desarrollo , Conejos , Radiografía , Espectrometría de Masas en Tándem , Tibia/diagnóstico por imagen , Tibia/microbiología , Regulación hacia Arriba
20.
Eur Cell Mater ; 27: 26-37; discussion 37-8, 2014 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-24464726

RESUMEN

Biomaterial implants and devices increase the risk of microbial infections due to the biofilm mode of growth of infecting bacteria on implant materials, in which bacteria are protected against antibiotic treatment and the local immune system. Matrix-metalloproteinases (MMPs) and cell surface integrin receptors facilitate transmigration of inflammatory cells toward infected or inflamed tissue. This study investigates the relationship between MMP- and integrin-expression and the clearance of infecting Staphylococcus aureus around implanted biomaterials in a murine model.MMP- and integrin αvß3-expression were monitored in mice, with and without subcutaneously implanted biomaterial samples, in the absence and presence of bioluminescent S. aureus Xen36. Staphylococcal persistence was imaged longitudinally over time using bioluminescence imaging. The activatable MMPSense®680 and integrin-targeted IntegriSense®750 probes were injected on different days after implantation and their signal intensity and localisation monitored using fluorescence imaging. After sacrifice 7 or 16 days post-implantation, staphylococci from biomaterial samples and surrounding tissues were cultured on agar-plates and presence of host inflammatory cells was histologically evaluated.MMP- and integrin-expression were equally enhanced in presence of staphylococci or biomaterials up to 7 days post-implantation, but their localisation along the biomaterial samples differed. Bacterial clearance from tissue was higher in the absence of biomaterials. It is of clinical relevance that MMP- and integrin-expression were enhanced in presence of both staphylococci and biomaterials, although the immune system in the presence of biomaterials remained hampered in eradicating bacteria during the first 7 days post-implantation.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Implantes Experimentales/microbiología , Integrina alfaVbeta3/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Infecciones Estafilocócicas/metabolismo , Animales , Integrina alfaVbeta3/genética , Metaloproteinasas de la Matriz/genética , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Infecciones Estafilocócicas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...