Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Nano Lett ; 23(17): 8326-8330, 2023 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-37611221

RESUMO

Bacterially induced sepsis requires rapid bacterial detection and identification. Hours count for critically ill septic patients, while current culture-based detection requires at least 10 h up to several days. Here, we apply a microfluidic device equipped with a bacterially activated, macrophage-membrane-coating on nanowired-Si adsorbent surfaces for rapid, bacterial detection and Gram-identification in bacterially contaminated blood. Perfusion of suspensions of Gram-negative or Gram-positive bacteria through a microfluidic device equipped with membrane-coated adsorbent surfaces detected low (<10 CFU/mL) bacterial levels. Subsequent, in situ fluorescence-staining yielded Gram-identification for guiding antibiotic selection. In mixed Escherichia coli and Staphylococcus aureus suspensions, Gram-negative and Gram-positive bacteria were detected in the same ratios as those fixed in suspension. Results were validated with a 100% correct score by blinded evaluation (two observers) of 15 human blood samples, spiked with widely different bacterial strains or combinations of strains, demonstrating the potential of the platform for rapid (1.5 h in total) diagnosis of bacterial sepsis.


Assuntos
Bactérias , Sepse , Humanos , Suspensões , Dispositivos Lab-On-A-Chip , Escherichia coli , Macrófagos , Sepse/diagnóstico
2.
Angew Chem Int Ed Engl ; 60(32): 17714-17719, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34028150

RESUMO

A lipid named DCPA was synthesized under microwave-assisted heating. DCPA possesses a pyridine betaine, hydrophilic group that can be complexed with water through hydrogen bonding (DCPA-H2 O). DCPA-H2 O liposomes became protonated relatively fast already at pH<6.8, due to the high HOMO binding energy of DCPA-H2 O. In murine models, DCPA-H2 O liposomes had longer blood circulation times than natural DPPC or cationic DCPM liposomes, while after tail-vein injection DCPA-H2 O liposomes targeted faster to solid tumors and intra-abdominal infectious biofilms. Therapeutic efficacy in a murine, infected wound-healing model of tail-vein injected ciprofloxacin-loaded DCPA-H2 O liposomes exceeded the ones of clinically applied ciprofloxacin as well as of ciprofloxacin-loaded DPPC or DCPM liposomes.


Assuntos
Portadores de Fármacos/farmacocinética , Lipossomos/farmacocinética , Neoplasias/diagnóstico por imagem , Infecções Estafilocócicas/diagnóstico por imagem , Água/química , Acetatos/síntese química , Acetatos/farmacocinética , Animais , Antibacterianos/uso terapêutico , Biofilmes , Ciprofloxacina/uso terapêutico , Portadores de Fármacos/síntese química , Feminino , Corantes Fluorescentes/química , Células Hep G2 , Humanos , Concentração de Íons de Hidrogênio , Lipossomos/química , Masculino , Camundongos Endogâmicos BALB C , Mycobacterium tuberculosis/fisiologia , Compostos de Piridínio/síntese química , Compostos de Piridínio/farmacocinética , Ratos Sprague-Dawley , Rodaminas/química , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/fisiopatologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/fisiologia , Tuberculose/diagnóstico por imagem , Tuberculose/fisiopatologia
3.
ACS Appl Mater Interfaces ; 12(31): 34610-34619, 2020 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-32633488

RESUMO

Photothermal nanoparticles locally release heat when irradiated by near-infrared (NIR). Clinical applications initially involved tumor treatment, but currently extend toward bacterial infection control. Applications toward much smaller, micrometer-sized bacterial infections, however, bear the risk of collateral damage by dissipating heat into tissues surrounding an infection site. This can become a complication when photothermal nanoparticle coatings are clinically applied on biomaterial surfaces requiring tissue integration, such as titanium-made, bone-anchored dental implants. Dental implants can fail due to infection in the pocket formed between the implant screw and the surrounding soft tissue ("peri-implantitis"). We address the hitherto neglected potential complication of collateral tissue damage by evaluating photothermal, polydopamine nanoparticle (PDA-NP) coatings on titanium surfaces in different coculture models. NIR irradiation of PDA-NP-coated (200 µg/cm2) titanium surfaces with adhering Staphylococcus aureus killed staphylococci within an irradiation time window of around 3 min. Alternatively, when covered with human gingival fibroblasts, this irradiation time window maintained surface coverage by fibroblasts. Contaminating staphylococci on PDA-NP-coated titanium surfaces, as can be per-operatively introduced, reduced surface coverage by fibroblasts, and this could be prevented by NIR irradiation for 5 min or longer prior to allowing fibroblasts to adhere and grow. Negative impacts of early postoperative staphylococcal challenges to an existing fibroblast layer covering a coated surface were maximally prevented by 3 min NIR irradiation. Longer irradiation times caused collateral fibroblast damage. Late postoperative staphylococcal challenges to a protective keratinocyte layer covering a fibroblast layer required 10 min NIR irradiation for adverting a staphylococcal challenge. This is longer than foreseen from monoculture studies because of additional heat uptake by the keratinocyte layer. Summarizing, photothermal treatment of biomaterial-associated infection requires precise timing of NIR irradiation to prevent collateral damage to tissues surrounding the infection site.


Assuntos
Antibacterianos/farmacologia , Indóis/farmacologia , Nanopartículas/química , Polímeros/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Temperatura , Titânio/farmacologia , Antibacterianos/química , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Fibroblastos/microbiologia , Humanos , Indóis/química , Testes de Sensibilidade Microbiana , Tamanho da Partícula , Processos Fotoquímicos , Polímeros/química , Propriedades de Superfície , Titânio/química
4.
ACS Biomater Sci Eng ; 6(2): 933-945, 2020 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33464836

RESUMO

Biomaterial-associated infections often arise from contaminating bacteria adhering to an implant surface that are introduced during surgical implantation and not effectively eradicated by antibiotic treatment. Whether or not infection develops from contaminating bacteria depends on an interplay between bacteria contaminating the biomaterial surface and tissue cells trying to integrate the surface with the aid of immune cells. The biomaterial surface plays a crucial role in defining the outcome of this race for the surface. Tissue integration is considered the best protection of a biomaterial implant against infectious bacteria. This paper aims to determine whether and how macrophages aid osteoblasts and human mesenchymal stem cells to adhere and spread over gold nanoparticle (GNP)-coatings with different hydrophilicity and roughness in the absence or presence of contaminating, adhering bacteria. All GNP-coatings had identical chemical surface composition, and water contact angles decreased with increasing roughness. Upon increasing the roughness of the GNP-coatings, the presence of contaminating Staphylococcus epidermidis in biculture with cells gradually decreased surface coverage by adhering and spreading cells, as in the absence of staphylococci. More virulent Staphylococcus aureus fully impeded cellular adhesion and spreading on smooth gold- or GNP-coatings, while Escherichia coli allowed minor cellular interaction. Murine macrophages in monoculture tended toward their pro-inflammatory "fighting" M1-phenotype on all coatings to combat the biomaterial, but in bicultures with contaminating, adhering bacteria, macrophages demonstrated Ym1 expression, indicative of polarization toward their anti-inflammatory "fix-and-repair" M2-phenotype. Damage repair of cells by macrophages improved cellular interactions on intermediately hydrophilic/rough (water contact angle 30 deg/surface roughness 118 nm) GNP-coatings in the presence of contaminating, adhering Gram-positive staphylococci but provided little aid in the presence of Gram-negative E. coli. Thus, the merits on GNP-coatings to influence the race for the surface and prevent biomaterial-associated infection critically depend on their hydrophilicity/roughness and the bacterial strain involved in contaminating the biomaterial surface.


Assuntos
Ouro , Macrófagos , Nanopartículas Metálicas , Animais , Adesão Celular , Movimento Celular , Escherichia coli , Humanos , Camundongos , Propriedades de Superfície
5.
Artigo em Inglês | MEDLINE | ID: mdl-30745390

RESUMO

Pseudomonas aeruginosa colonizes the sputum of most adult cystic fibrosis patients, forming difficult-to-eradicate biofilms in which bacteria are protected in their self-produced extracellular polymeric substance (EPS) matrices. EPS provide biofilms with viscoelastic properties, causing time-dependent relaxation after stress-induced deformation, according to multiple characteristic time constants. These time constants reflect different biofilm (matrix) components. Since the viscoelasticity of biofilms has been related to antimicrobial penetration but not yet bacterial killing, this study aims to relate killing of P. aeruginosa, in its biofilm mode of growth, by three antimicrobials to biofilm viscoelasticity. P. aeruginosa biofilms were grown for 18 h in a constant-depth film fermenter, with mucin-containing artificial sputum medium (ASM+), artificial sputum medium without mucin (ASM-), or Luria-Bertani (LB) broth; this yielded 100-µm-thick biofilms that differed in their amounts of matrix environmental DNA (eDNA) and polysaccharides. Low-load compression testing, followed by three-element Maxwell analyses, showed that the fastest relaxation component, associated with unbound water, was most important in LB-medium-grown biofilms. Slower components due to water with dissolved polysaccharides, insoluble polysaccharides, and eDNA were most important in the relaxation of ASM+-grown biofilms. ASM--grown biofilms showed intermediate stress relaxation. P. aeruginosa in LB-medium-grown biofilms was killed most by exposure to tobramycin, colistin, or an antimicrobial peptide, while ASM+ provided the most protective matrix, with less water and most insoluble polysaccharides and eDNA. In conclusion, stress relaxation of P. aeruginosa biofilms grown in different media revealed differences in matrix composition that, within the constraints of the antimicrobials and growth media applied, correlated with the matrix protection offered against different antimicrobials.


Assuntos
Antibacterianos/farmacologia , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Colistina/farmacologia , Meios de Cultura/química , Matriz Extracelular/química , Testes de Sensibilidade Microbiana , Mucinas , Pseudomonas aeruginosa/fisiologia , Tobramicina/farmacologia , Viscosidade
6.
J Orthop Res ; 37(2): 271-287, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30667561

RESUMO

Orthopedic device-related infection (ODRI), including both fracture-related infection (FRI) and periprosthetic joint infection (PJI), remain among the most challenging complications in orthopedic and musculoskeletal trauma surgery. ODRI has been convincingly shown to delay healing, worsen functional outcome and incur significant socio-economic costs. To address this clinical problem, ever more sophisticated technologies targeting the prevention and/or treatment of ODRI are being developed and tested in vitro and in vivo. Among the most commonly described innovations are antimicrobial-coated orthopedic devices, antimicrobial-loaded bone cements and void fillers, and dual osteo-inductive/antimicrobial biomaterials. Unfortunately, translation of these technologies to the clinic has been limited, at least partially due to the challenging and still evolving regulatory environment for antimicrobial drug-device combination products, and a lack of clarity in the burden of proof required in preclinical studies. Preclinical in vivo testing (i.e. animal studies) represents a critical phase of the multidisciplinary effort to design, produce and reliably test both safety and efficacy of any new antimicrobial device. Nonetheless, current in vivo testing protocols, procedures, models, and assessments are highly disparate, irregularly conducted and reported, and without standardization and validation. The purpose of the present opinion piece is to discuss best practices in preclinical in vivo testing of antimicrobial interventions targeting ODRI. By sharing these experience-driven views, we aim to aid others in conducting such studies both for fundamental biomedical research, but also for regulatory and clinical evaluation. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:271-287, 2019.


Assuntos
Experimentação Animal/normas , Fixação Interna de Fraturas/efeitos adversos , Fixadores Internos/efeitos adversos , Infecções Relacionadas à Prótese , Animais , Anti-Infecciosos/administração & dosagem , Modelos Animais , Infecções Relacionadas à Prótese/microbiologia , Projetos de Pesquisa
7.
Front Chem ; 7: 861, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31970146

RESUMO

Cascade reactions integrate two or more reactions, of which each subsequent reaction can only start when the previous reaction step is completed. Employing natural substrates in the human body such as glucose and oxygen, cascade reactions can generate reactive oxygen species (ROS) to kill tumor cells, but cascade reactions may also have potential as a direly needed, novel bacterial infection-control strategy. ROS can disintegrate the EPS matrix of infectious biofilm, disrupt bacterial cell membranes, and damage intra-cellular DNA. Application of cascade reactions producing ROS as a new infection-control strategy is still in its infancy. The main advantages for infection-control cascade reactions include the fact that they are non-antibiotic based and induction of ROS resistance is unlikely. However, the amount of ROS generated is generally low and antimicrobial efficacies reported are still far <3-4 log units necessary for clinical efficacy. Increasing the amounts of ROS generated by adding more substrate bears the risk of collateral damage to tissue surrounding an infection site. Collateral tissue damage upon increasing substrate concentrations may be prevented by locally increasing substrate concentrations, for instance, using smart nanocarriers. Smart, pH-responsive nanocarriers can self-target and accumulate in infectious biofilms from the blood circulation to confine ROS production inside the biofilm to yield long-term presence of ROS, despite the short lifetime (nanoseconds) of individual ROS molecules. Increasing bacterial killing efficacies using cascade reaction components containing nanocarriers constitutes a first, major challenge in the development of infection-control cascade reactions. Nevertheless, their use in combination with clinical antibiotic treatment may already yield synergistic effects, but this remains to be established for cascade reactions. Furthermore, specific patient groups possessing elevated levels of endogenous substrate (for instance, diabetic or cancer patients) may benefit from the use of cascade reaction components containing nanocarriers.

8.
Chem Sci ; 9(21): 4730-4735, 2018 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-29910923

RESUMO

Single cell surface engineering provides the most efficient, non-genetic strategy to enhance cell stability. However, it remains a huge challenge to improve cell stability in complex artificial environments. Here, a soft biohybrid interfacial layer is fabricated on individual living-cell surfaces by their exposure to a suspension of gold nanoparticles and l-cysteine to form a protecting functional layer to which porous silica layers were bound yielding pores with a diameter of 3.9 nm. The living cells within the bilayered nanoshells maintained high viability (96 ± 2%) as demonstrated by agar plating, even after five cycles of simultaneous exposure to high temperature (40 °C), lyticase and UV light. Moreover, yeast cells encapsulated in bilayered nanoshells were more recyclable than native cells due to nutrient storage in the shell.

9.
Biomacromolecules ; 19(6): 2023-2033, 2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29584416

RESUMO

Inadvertent photosensitizer-activation and singlet-oxygen generation hampers clinical application of photodynamic therapies of superficial tumors or subcutaneous infections. Therefore, a reversible photoswitchable system was designed in micellar nanocarriers using ZnTPP as a photosensitizer and BDTE as a photoswitch. Singlet-oxygen generation upon irradiation didnot occur in closed-switch micelles with ZnTPP/BDTE feeding ratios >1:10. Deliberate switch closure/opening within 65-80 min was possible through thin layers of porcine tissue in vitro, increasing for thicker layers. Inadvertent opening of the switch by simulated daylight, took several tens of hours. Creating deliberate cell damage and prevention of inadvertent damage in vitro and in mice could be done at lower ZnTPP/BDTE feeding ratios (1:5) in open-switch micelles and at higher irradiation intensities than inferred from chemical clues to generate singlet-oxygen. The reduction of inadvertent photosensitizer activation in micellar nanocarriers, while maintaining the ability to kill tumor cells and infectious bacteria established here, brings photodynamic therapies closer to clinical application.


Assuntos
Nanoestruturas/química , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Oxigênio Singlete/metabolismo , Células 3T3 , Animais , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Espectroscopia de Ressonância de Spin Eletrônica , Feminino , Células HeLa , Humanos , Lactonas/química , Camundongos , Camundongos Endogâmicos BALB C , Micelas , Fármacos Fotossensibilizantes/administração & dosagem , Polietilenoglicóis/química , Porfirinas/química , Oxigênio Singlete/química , Espectrofotometria Ultravioleta , Zinco/química
10.
Biofouling ; 33(9): 712-721, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28868925

RESUMO

Transmission is a main route for bacterial contamination, involving bacterial detachment from a donor and adhesion to receiver surfaces. This work aimed to compare transmission of an extracellular polymeric substance (EPS) producing and a non-EPS producing Staphylococcus epidermidis strain from biofilms on stainless steel. After transmission, donor surfaces remained fully covered with biofilm, indicating transmission through cohesive failure in the biofilm. Counter to the numbers of biofilm bacteria, the donor and receiver biofilm thicknesses did not add up to the pre-transmission donor biofilm thickness, suggesting more compact biofilms after transmission, especially for non-EPS producing staphylococci. Accordingly, staphylococcal density per unit biofilm volume had increased from 0.20 to 0.52 µm-3 for transmission of the non-EPS producing strain under high contact pressure. The EPS producing strain had similar densities before and after transmission (0.17 µm-3). This suggests three phases in biofilm transmission: (1) compression, (2) separation and (3) relaxation of biofilm structure to its pre-transmission density in EPS-rich biofilms.


Assuntos
Aderência Bacteriana , Biofilmes/crescimento & desenvolvimento , Aço Inoxidável , Staphylococcus epidermidis/crescimento & desenvolvimento , Microscopia Confocal , Pressão , Staphylococcus epidermidis/fisiologia , Propriedades de Superfície , Tomografia de Coerência Óptica
11.
Microb Biotechnol ; 10(6): 1744-1752, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28771954

RESUMO

In real-life situations, bacteria are often transmitted from biofilms growing on donor surfaces to receiver ones. Bacterial transmission is more complex than adhesion, involving bacterial detachment from donor and subsequent adhesion to receiver surfaces. Here, we describe a new device to study shear-induced bacterial transmission from a (stainless steel) pipe to a (silicone rubber) tube and compare transmission of EPS-producing and non-EPS-producing staphylococci. Transmission of an entire biofilm from the donor to the receiver tube did not occur, indicative of cohesive failure in the biofilm rather than of adhesive failure at the donor-biofilm interface. Biofilm was gradually transmitted over an increasing length of receiver tube, occurring mostly to the first 50 cm of the receiver tube. Under high-shearing velocity, transmission of non-EPS-producing bacteria to the second half decreased non-linearly, likely due to rapid thinning of the lowly lubricious biofilm. Oppositely, transmission of EPS-producing strains to the second tube half was not affected by higher shearing velocity due to the high lubricity and stress relaxation of the EPS-rich biofilms, ensuring continued contact with the receiver. The non-linear decrease of ongoing bacterial transmission under high-shearing velocity is new and of relevance in for instance, high-speed food slicers and food packaging.


Assuntos
Biofilmes , Elastômeros de Silicone/química , Aço Inoxidável/química , Staphylococcus/fisiologia , Aderência Bacteriana , Staphylococcus/química
12.
ACS Appl Mater Interfaces ; 9(13): 12118-12129, 2017 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-28291321

RESUMO

Bacterial adhesion and biofilm formation on surfaces are troublesome in many industrial processes. Here, nanoporous and nanopillared aluminum surfaces were engineered by anodizing and postetching processes and made hydrophilic (using the inherent oxide layer) or hydrophobic (applying a Teflon coating) with the aim of discouraging bacterial adhesion. Adhesion of Staphylococcus aureus ATCC 12600 (Gram-positive, spherically shaped) and Escherichia coli K-12 (Gram-negative, rod-shaped) was evaluated to the nanoengineered surfaces under both static and flow conditions (fluid shear rate of 37 s-1). Compared to a nonstructured electropolished flat surface, the nanostructured surfaces significantly reduced the number of adhering colony forming units (CFUs) for both species, as measured using agar plating. For the hydrophilic surfaces, this was attributed to a decreased contact area, reducing bacterial adhesion forces on nanoporous and nanopillared surfaces to 4 and 2 nN, respectively, from 8 nN on flat surfaces. Reductions in the numbers of adhering CFUs were more marked on hydrophobic surfaces under flow, amounting to more than 99.9% and 99.4% for S. aureus and E. coli on nanopillared surfaces, respectively. Scanning electron microscopy revealed a few bacteria found on the hydrophobic nanopillared surfaces adhered predominantly to defective or damaged areas, whereas the intact area preserving the original nanopillared morphology was virtually devoid of adhering bacteria. The greater decrease in bacterial adhesion to hydrophobic nanopillared surfaces than to hydrophilic or nanoporous ones is attributed to effective air entrapment in the three-dimensional pillar morphology, rendering them superhydrophobic and slippery, in addition to providing a minimized contact area for bacteria to adhere to.


Assuntos
Aderência Bacteriana , Alumínio , Escherichia coli , Escherichia coli K12 , Interações Hidrofóbicas e Hidrofílicas , Staphylococcus aureus , Propriedades de Superfície
13.
Pathog Dis ; 74(4): ftw029, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27060097

RESUMO

ITALIC! Staphylococcus aureusbiofilms can be found on vaginal epithelia, secreting toxins and causing inflammation. The co-vaginal species ITALIC! Lactobacilluscan alter staphylococcal-induced epithelial secretion of inflammatory cytokines and quench staphylococcal toxic shock syndrome toxin-1 secretion. It is hypothesized that these effects of lactobacilli require direct physical contact between lactobacilli, staphylococci and the epithelium. Indeed, lactobacilli only reduced ITALIC! S. aureus-induced inflammatory cytokine expression when allowed physical contact with vaginal epithelial cells. Furthermore, a reduction in toxic shock syndrome toxin-1 secretion only occurred when a probiotic ITALIC! Lactobacillusstrain was allowed contact, but not when being physically separated from ITALIC! S. aureus Bacterial-probe atomic force microscopy demonstrated that lactobacilli and staphylococci strongly adhere to epithelial cells, while lactobacilli adhere stronger to staphylococci than staphylococci to each other, giving lactobacilli opportunity to penetrate and reside in staphylococcal biofilms, as visualized using confocal laser scanning microscopy with fluorescence ITALIC! in situhybridization probes. These results identify that physical contact and biochemical signaling by lactobacilli are intrinsically linked mechanisms that reduce virulence of ITALIC! S. aureusbiofilm.


Assuntos
Enterotoxinas/metabolismo , Lactobacillus/fisiologia , Interações Microbianas , Staphylococcus aureus/fisiologia , Aderência Bacteriana , Toxinas Bacterianas , Linhagem Celular , Citocinas/metabolismo , Células Epiteliais , Feminino , Humanos , Mediadores da Inflamação/metabolismo , Superantígenos , Vaginose Bacteriana/metabolismo , Vaginose Bacteriana/microbiologia
14.
Cell Microbiol ; 18(4): 605-14, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26477544

RESUMO

Vaginal epithelium is colonized by different bacterial strains and species. The bacterial composition of vaginal biofilms controls the balance between health and disease. Little is known about the relative contribution of the epithelial and bacterial cell surfaces to bacterial adhesion and whether and how adhesion is regulated over cell membrane regions. Here, we show that bacterial adhesion forces with cell membrane regions not located above the nucleus are stronger than with regions above the nucleus both for vaginal pathogens and different commensal and probiotic lactobacillus strains involved in health. Importantly, adhesion force ratios over membrane regions away from and above the nucleus coincided with the ratios between numbers of adhering bacteria over both regions. Bacterial adhesion forces were dramatically decreased by depleting the epithelial cell membrane of cholesterol or sub-membrane cortical actin. Thus, epithelial cells can regulate membrane regions to which bacterial adhesion is discouraged, possibly to protect the nucleus.


Assuntos
Aderência Bacteriana , Membrana Celular/fisiologia , Células Epiteliais/microbiologia , Vagina/microbiologia , Adesividade , Linhagem Celular , Feminino , Humanos
15.
Int J Oral Sci ; 7(4): 250-8, 2015 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-26674427

RESUMO

Sub-gingival anaerobic pathogens can colonize an implant surface to compromise osseointegration of dental implants once the soft tissue seal around the neck of an implant is broken. In vitro evaluations of implant materials are usually done in monoculture studies involving either tissue integration or bacterial colonization. Co-culture models, in which tissue cells and bacteria battle simultaneously for estate on an implant surface, have been demonstrated to provide a better in vitro mimic of the clinical situation. Here we aim to compare the surface coverage by U2OS osteoblasts cells prior to and after challenge by two anaerobic sub-gingival pathogens in a co-culture model on differently modified titanium (Ti), titanium-zirconium (TiZr) alloys and zirconia surfaces. Monoculture studies with either U2OS osteoblasts or bacteria were also carried out and indicated significant differences in biofilm formation between the implant materials, but interactions with U2OS osteoblasts were favourable on all materials. Adhering U2OS osteoblasts cells, however, were significantly more displaced from differently modified Ti surfaces by challenging sub-gingival pathogens than from TiZr alloys and zirconia variants. Combined with previous work employing a co-culture model consisting of human gingival fibroblasts and supra-gingival oral bacteria, results point to a different material selection to stimulate the formation of a soft tissue seal as compared to preservation of osseointegration under the unsterile conditions of the oral cavity.


Assuntos
Implantes Dentários/microbiologia , Materiais Dentários/química , Osseointegração/fisiologia , Osteoblastos/fisiologia , Porphyromonas gingivalis/fisiologia , Prevotella intermedia/fisiologia , Condicionamento Ácido do Dente/métodos , Ligas/química , Aderência Bacteriana/fisiologia , Técnicas Bacteriológicas , Biofilmes , Adesão Celular/fisiologia , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Cerâmica/química , Técnicas de Cocultura , Ligas Dentárias/química , Corrosão Dentária/métodos , Polimento Dentário/métodos , Humanos , Propriedades de Superfície , Titânio/química , Ítrio/química , Zircônio/química
16.
ACS Appl Mater Interfaces ; 7(36): 20304-13, 2015 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-26305913

RESUMO

Titanium is often applied in implant surgery, but frequently implicated in infections associated with bacterial adhesion and growth on the implant surface. Here, we show that hierarchical nanostructuring of titanium and the subsequent coating of resulting topographical features with a self-defensive, antibacterial layer-by-layer (LbL) film enables a synergistic action of hierarchical nanotopography and localized, bacteria-triggered antibiotic release to dramatically enhance the antibacterial efficiency of surfaces. Although sole nanostructuring of titanium substrates did not significantly affect adhesion and growth of Staphylococcus aureus, the coating of 3D-nanopillared substrates with an ultrathin tannic acid/gentamicin (TA/G) LbL film resulted in a 10-fold reduction of the number of surface-attached bacteria. This effect is attributed to the enlarged surface area of the nanostructured coating available for localized bacteria-triggered release of antibiotics, as well as to the lower bacterial adhesion forces resulting in subsided activation of bacterial antibiotic-defense mechanisms when bacteria land on nanopillar tips. The result shows that a combination of 3D nanostructuring with a bacteria-triggered antibiotic-releasing coating presents a unique way to dramatically enhance antibacterial efficacy of biomaterial implants.


Assuntos
Antibacterianos/química , Materiais Revestidos Biocompatíveis/química , Nanoestruturas/química , Titânio/química , Antibacterianos/farmacologia , Aderência Bacteriana/efeitos dos fármacos , Materiais Revestidos Biocompatíveis/farmacologia , Gentamicinas/química , Gentamicinas/farmacologia , Microscopia de Força Atômica , Microscopia de Fluorescência , Staphylococcus aureus/efeitos dos fármacos , Taninos/química
17.
J Biomed Mater Res A ; 103(11): 3590-8, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25966819

RESUMO

Bacterial contamination during biomaterial implantation is often unavoidable, yielding a combat between cells and bacteria. Here we aim to determine the modulatory function of bacterial components on stem-cell, fibroblast, and osteoblast adhesion to a titanium alloy, including the role of toll-like-receptors (TLRs). Presence of heat-sacrificed Staphylococcus epidermidis, Staphylococcus aureus, Escherichia coli, or Pseudomonas aeruginosa induced dose and cell-type dependent responses. Stem-cells were most sensitive to bacterial presence, demonstrating decreased adhesion number yet increased adhesion effort with a relatively large focal adhesion contact area. Blocking TLRs had no effect on stem-cell adhesion in presence of S. aureus, but blocking both TLR2 and TLR4 induced an increased adhesion effort in presence of E. coli. Neither lipopolysaccharide, lipoteichoic acid, nor bacterial DNA provoked the same cell response as did whole bacteria. Herewith we suggest a new mechanism as to how biomaterials are integrated by cells despite the unavoidable presence of bacterial contamination. Stimulation of host cell integration of implant surfaces may open a new window to design new biomaterials with enhanced healing, thereby reducing the risk of biomaterial-associated infection of both "hardware-based" implants as well as of tissue-engineered constructs, known to suffer from similarly high infection risks as currently prevailing in "hardware-based" implants.


Assuntos
Bactérias/efeitos dos fármacos , Materiais Biocompatíveis/farmacologia , Próteses e Implantes , Ligas/farmacologia , Adesão Celular/efeitos dos fármacos , Contagem de Células , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Imunofluorescência , Adesões Focais/efeitos dos fármacos , Adesões Focais/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Titânio/farmacologia , Receptores Toll-Like/metabolismo
18.
Acta Biomater ; 18: 1-8, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25752975

RESUMO

Biomaterial-associated-infection causes failure of biomaterial implants. Many new biomaterials have been evaluated for their ability to inhibit bacterial colonization and stimulate tissue-cell-integration, but neglect the role of immune cells. This paper compares macrophage phagocytosis of adhering Staphylococcus aureus on cationic-coatings and patterned poly(ethylene)glycol-hydrogels versus common biomaterials and stainless steel in order to identify surface conditions that promote clearance of adhering bacteria. Staphylococci were allowed to adhere and grow on the materials in a parallel-plate-flow-chamber, after which murine macrophages were introduced. From the decrease in the number of adhering staphylococci, phagocytosis-rates were calculated, and total macrophage displacements during an experiment determined. Hydrophilic surfaces had the lowest phagocytosis-rates, while common biomaterials had intermediate phagocytosis-rates. Patterning of poly(ethylene)glycol-hydrogel coatings increased phagocytosis-rates to the level of common biomaterials, while on cationic-coatings phagocytosis-rates remained relatively low. Likely, phagocytosis-rates on cationic coatings are hampered relative to common biomaterials through strong electrostatic binding of negatively-charged macrophages and staphylococci. On polymeric biomaterials and glass, phagocytosis-rates increased with macrophage displacement, while both parameters increased with biomaterial surface hydrophobicity. Thus hydrophobicity is a necessary surface condition for effective phagocytosis. Concluding, next-generation biomaterials should account for surface effects on phagocytosis in order to enhance the ability of these materials to resist biomaterial-associated-infection.


Assuntos
Aderência Bacteriana/efeitos dos fármacos , Materiais Revestidos Biocompatíveis/farmacologia , Hidrogel de Polietilenoglicol-Dimetacrilato/farmacologia , Macrófagos/citologia , Fagocitose/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos , Animais , Cátions , Linhagem Celular , Contagem de Colônia Microbiana , Camundongos , Polímeros/farmacologia
19.
Int J Oral Sci ; 7(1): 42-8, 2015 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-25572920

RESUMO

Retention wires permanently bonded to the anterior teeth are used after orthodontic treatment to prevent the teeth from relapsing to pre-treatment positions. A disadvantage of bonded retainers is biofilm accumulation on the wires, which produces a higher incidence of gingival recession, increased pocket depth and bleeding on probing. This study compares in vivo biofilm formation on single-strand and multi-strand retention wires with different oral health-care regimens. Two-centimetre wires were placed in brackets that were bonded to the buccal side of the first molars and second premolars in the upper arches of 22 volunteers. Volunteers used a selected toothpaste with or without the additional use of a mouthrinse containing essential oils. Brushing was performed manually. Regimens were maintained for 1 week, after which the wires were removed and the oral biofilm was collected to quantify the number of organisms and their viability, determine the microbial composition and visualize the bacteria by electron microscopy. A 6-week washout period was employed between regimens. Biofilm formation was reduced on single-strand wires compared with multi-strand wires; bacteria were observed to adhere between the strands. The use of antibacterial toothpastes marginally reduced the amount of biofilm on both wire types, but significantly reduced the viability of the biofilm organisms. Additional use of the mouthrinse did not result in significant changes in biofilm amount or viability. However, major shifts in biofilm composition were induced by combining a stannous fluoride- or triclosan-containing toothpaste with the mouthrinse. These shifts can be tentatively attributed to small changes in bacterial cell surface hydrophobicity after the adsorption of the toothpaste components, which stimulate bacterial adhesion to the hydrophobic oil, as illustrated for a Streptococcus mutans strain.


Assuntos
Biofilmes , Higiene Bucal , Contenções Ortodônticas , Aço Inoxidável , Sequência de Bases , Primers do DNA , Eletroforese em Gel de Poliacrilamida , Humanos , Reação em Cadeia da Polimerase
20.
Biomaterials ; 35(21): 5446-52, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24726539

RESUMO

Surfaces with cell adhesiveness modulated at micro length scales can exploit differences between tissue/bacterial cell size, membrane/wall plasticity, and adhesion mechanisms to differentially control tissue-cell/material and bacteria/material interactions. This study explores the short-term interactions of Staphylococcus aureus and osteoblast-like cells with surfaces consisting of cell-adhesive circular patches (1-5 µm diameter) separated by non-adhesive electron-beam patterned poly(ethylene glycol) hydrogel thin films at inter-patch distances of 0.5-10 µm. Osteoblast-like U2OS cells both bind to and spread on the modulated surfaces, in some cases when the cell-adhesive area comprises only 9% of the total surface and in several cases at least as well as on the continuously adhesive control surfaces. In contrast, S. aureus adhesion rates are 7-20 times less on the modulated surfaces than on the control surfaces. Furthermore, the proliferation of those bacteria that do adhere is inhibited by the lateral confinement imposed by the non-adhesive boundaries surrounding each patch. These findings suggest a new approach to create biomaterial surfaces that may promote healing while simultaneously reducing the probability of infection.


Assuntos
Materiais Biocompatíveis/química , Biofilmes/crescimento & desenvolvimento , Aderência Bacteriana , Adesão Celular , Linhagem Celular Tumoral , Humanos , Hidrogéis/química , Osteoblastos/metabolismo , Osteoblastos/microbiologia , Polietilenoglicóis/química , Staphylococcus aureus/crescimento & desenvolvimento , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA