Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 506
Filtrar
1.
Biomaterials ; 293: 121957, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36549042

RESUMO

Aging population has been boosting the need for orthopedic implants. However, biofilm has been a major obstacle for orthopedic implants due to its insensitivity to antibiotics and tendency to drive antimicrobial resistance. Herein, an antibacterial polypeptide coating with excellent in vivo adhesive capacity was prepared to prevent implants from forming biofilms and inducing acquired antibiotic resistance. A peptide-based copolymer, poly[phenylalanine10-stat-lysine12]-block-3,4-dihydroxy-l-phenylalanine [Poly(Phe10-stat-Lys12)-DOPA] was modularly designed, where poly(Phe10-stat-Lys12) is antibacterial polypeptide with high antibacterial activity, and DOPA provides strong adhesion in both wet and dry microenvironments. Meanwhile, compared to traditional "graft-onto" methods, this antibacterial coating can be facilely achieved by immersing Titanium substrates into antibacterial polypeptide solution for 5 min at room temperature. The poly(Phe10-stat-Lys12)-DOPA polymer showed good antibacterial activity with minimum inhibitory concentrations against S. aureus and E. coli of 32 and 400 µg/mL, respectively. Compared to obvious antimicrobial resistance of S. aureus after continuous treatment with vancomycin, this antibacterial coating doesn't drive antimicrobial resistance upon long-term utilization. Transcriptome sequencing and qPCR tests further confirmed that the antibacterial coating was able to inhibit the expression of multiple peptide resistance factor (mprF) and lipoteichoic acid modification D-alanylation genes (dltB and dltC) that can increase the net positive charge of bacterial cell wall to induce the resistance to cationic antimicrobial peptides. In vivo experiments confirmed that this poly(Phe10-stat-Lys12)-DOPA coating can both effectively prevent biofilm formation through surface contact sterilization and avoid local and systemic infections. Overall, we proposed a facile method for preparing antibacterial orthopedic implants with longer indwelling time and without inducing antimicrobial resistance by coating a polypeptide-based polymer on the implants.


Assuntos
Antibacterianos , Peptídeos Catiônicos Antimicrobianos , Biofilmes , Materiais Revestidos Biocompatíveis , Titânio , Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Biofilmes/efeitos dos fármacos , Materiais Revestidos Biocompatíveis/farmacologia , Di-Hidroxifenilalanina/farmacologia , Escherichia coli , Polímeros/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Titânio/farmacologia , Próteses e Implantes/microbiologia , Farmacorresistência Bacteriana
2.
Rev. bras. ortop ; 57(4): 689-696, Jul.-Aug. 2022. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1394867

RESUMO

Abstract Objective To evaluate the sensitivity and specificity of the quantitative real-time polymerase chain reaction (qPCR) for 16S rDNA gene screening using sonicated fluid from orthopedic implants. Methods A retrospective study was conducted on 73 sonicated fluids obtained from patients with infection associated with orthopedic implants. The samples were subjected to conventional culture and molecular testing using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) and qPCR for 16S rDNA. The cycle threshold values were used to define a cut-off of the qPCR of the 16S rDNA for negative and positive cultures. Results No statistical differences were observed between the positive and negative culture groups based on the time from the first surgery to infection (p= 0.958), age (p =0.269), or general comorbidities. Nevertheless, a statistical difference was found between the mean duration of antibiotic use before device removal (3.41 versus 0.94; p =0.016). Bacterial DNA was identified in every sample from the sonicated fluids. The median cycle thresholds of the positive and negative cultures were of 25.6 and 27.3 respectively (p< 0.001). As a diagnostic tool, a cycle threshold cut-off of 26.89 demonstrated an area under the curve of the receiver operating characteristic of 0.877 (p≤ 0.001). Conclusion The presence of antimicrobial agents for more than 72 hours decreased culture positivity, but did not influence the qPCR results. Despite this, amplification of the 16S rDNA may overestimate infection diagnosis.


Resumo Objetivo Avaliar a sensibilidade e a especificidade da reação em cadeia de polimerase em tempo real quantitativa (quantitative real-time polymerase chain reaction, qPCR, em inglês) para a triagem do gene rDNA 16S, com a utilização do fluido sonicado de implantes ortopédicos. Métodos Um estudo retrospectivo foi realizado em 73 fluidos sonicados obtidos de pacientes com infecção associada aos implantes ortopédicos. As amostras foram submetidas a cultura convencional e a teste molecular utilizando ionização e dessorção a laser assistida por matriz com espectrometria de massa por tempo de voo (matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, MALDI-TOF MS, em inglês) e qPCR para o gene rDNA 16S. Os valores limiares do ciclo foram usados para definir um ponto de corte para a qPCR do gene rDNA 16S para culturas negativas e positivas. Resultados Não foram observadas diferenças estatísticas entre os grupos de cultura positiva e negativa com base no tempo desde a primeira cirurgia até a infecção (p= 0,958), na idade (p= 0,269), ou nas comorbidades em geral. No entanto, uma diferença estatística foi encontrada entre a duração média do uso de antibióticos antes da remoção do dispositivo (3,41 versus 0,94; p= 0,016). O DNA bacteriano foi identificado em todas as amostras dos fluidos sonicados. Os limiares do ciclo médio de culturas positivas e negativas foram de 25,6 e 27,3, respectivamente (p< 0,001). Como uma ferramenta de diagnóstico, um corte do limite do ciclo de 26,89 demonstrou uma área sob a curva da característica de operação do receptor de 0,877 (p ≤ 0,001). Conclusão A presença de agentes antimicrobianos por mais de 72 horas diminuiu a positividade da cultura, mas não influenciou os resultados da qPCR. Apesar disso, a amplificação do rDNA 16S pode sobrestimar o diagnóstico de infecção.


Assuntos
Humanos , Próteses e Implantes/microbiologia , Sonicação , Reação em Cadeia da Polimerase , Estudos Retrospectivos , Controle de Infecções , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Anti-Infecciosos
3.
Microbiol Spectr ; 10(1): e0161221, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35171021

RESUMO

Tranexamic acid (TXA) is extensively used in orthopedic surgery and traumatology as an antifibrinolytic agent to control intra- and postoperative bleeding and, therefore, indirectly, to reduce postsurgery infection rates. The hypothesis of an additional antibiotic effect against microorganisms associated with periprosthetic joint infection needs to be further evaluated. We aimed to assess whether TXA could reduce bacterial growth using an in vitro model. ATCC and clinical strains of staphylococci and Cutibacterium acnes were tested against TXA in both planktonic and sessile forms. We recorded the percent reduction in the following variables: log CFU/mL by microbiological culture, percentage of live cells by confocal laser scanning microscopy, and, additionally in sessile cells, metabolic activity by the 2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide salt (XTT) assay. Variables were compared between groups using the Kruskal-Wallis test, and the results were reported as median (interquartile range [IQR]). Statistical significance was set at a P value of <0.05. Clinical significance was defined as a reduction of ≥25%. TXA at 50 mg/mL led to a slight reduction in CFU counts (4.5%). However, it was at 10 mg/mL that the reduction reached 27.2% and 33.0% for log CFU/mL counts and percentage of live cells, respectively. TXA was not efficacious for reducing preformed 24-h mature staphylococci and 48-h mature C. acnes biofilms, regardless of its concentration. TXA did not exert an antimicrobial effect against bacterial biofilms. However, when bacteria were in the planktonic form, it led to a clinically and statistically significant reduction in bacterial growth at 10 mg/mL. IMPORTANCE The possible use of TXA as an antibiotic agent in addition to its antifibrinolytic effect may play an important role in the prevention of prosthetic joint infection.


Assuntos
Antibacterianos/farmacologia , Infecções por Bactérias Gram-Positivas/microbiologia , Propionibacteriaceae/efeitos dos fármacos , Próteses e Implantes/microbiologia , Infecções Relacionadas à Prótese/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus/efeitos dos fármacos , Ácido Tranexâmico/farmacologia , Biofilmes/efeitos dos fármacos , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Humanos , Testes de Sensibilidade Microbiana , Propionibacteriaceae/crescimento & desenvolvimento , Infecções Relacionadas à Prótese/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus/crescimento & desenvolvimento
4.
Eur J Clin Microbiol Infect Dis ; 41(1): 87-97, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34599708

RESUMO

Prosthetic joint infection (PJI) is a feared and challenging to diagnose complication after arthroplasty, with Staphylococcus epidermidis as the major pathogen. One important criteria to define PJI is the detection of phenotypically indistinguishable microorganisms with identical antibiotic susceptibility pattern in at least two different samples. However, owing to phenotypical variation within genetic clones and clonal variation within a phenotype, the criteria may be ambiguous. We investigated the extent of diversity among coagulase-negative staphylococci (CoNS) in PJI and characterised S. epidermidis isolates from PJI samples, specifically multiple S. epidermidis isolates identified in individual PJI patients. We performed a retrospective cohort study on 62 consecutive patients with PJI caused by CoNS from two hospitals in Northern Sweden. In 16/62 (26%) PJIs, multiple S. epidermidis isolates were available for whole-genome analyses. Hospital-adapted multidrug-resistant genetic clones of S. epidermidis were identified in samples from 40/62 (65%) of the patients using a combination of pulsed-field gel electrophoresis and multilocus sequence typing. Whole-genome sequencing showed the presence of multiple sequence types (STs) in 7/16 (44%) PJIs where multiple S. epidermidis isolates were available. Within-patient phenotypical variation in the antibiotic susceptibility and/or whole-genome antibiotic resistance gene content was frequent (11/16, 69%) among isolates with the same ST. The results highlight the ambiguity of S. epidermidis phenotypic characterisation as a diagnostic method in PJI and call for larger systematic studies for determining the frequency of CoNS diversity in PJIs, the implications of such diversity for microbiological diagnostics, and the therapeutic outcomes in patients.


Assuntos
Articulações/microbiologia , Infecções Relacionadas à Prótese/microbiologia , Staphylococcus epidermidis/fisiologia , Idoso , Antibacterianos/farmacologia , Farmacorresistência Bacteriana Múltipla , Feminino , Humanos , Articulações/cirurgia , Masculino , Testes de Sensibilidade Microbiana , Pessoa de Meia-Idade , Próteses e Implantes/microbiologia , Infecções Relacionadas à Prótese/tratamento farmacológico , Estudos Retrospectivos , Staphylococcus epidermidis/efeitos dos fármacos , Staphylococcus epidermidis/genética , Staphylococcus epidermidis/isolamento & purificação
6.
Bull Exp Biol Med ; 172(1): 46-48, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34792719

RESUMO

Comparison of activity of antibiotics against Staphylococcus aureus strains in their plankton form and in biofilms of varying maturity showed that the concentrations of antibiotics causing death of 90% S. aureus strains (MIC90) in their plankton form was 0.07-2.80 µg/ml and significantly (p<0.05) differed from MIC90 concentrations (3-245 µg/ml) for the strains in 24 and 48 h biofilms. MIC90 value was affected by the degree of biofilm maturity: microbial resistance in 48-h biofilms for all analyzed antibiotics was significantly (p<0.05) higher than in 24-h biofilms.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Infecções Relacionadas à Prótese/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Ciprofloxacina/farmacologia , Farmacorresistência Bacteriana Múltipla/fisiologia , Fluoroquinolonas/farmacologia , Testes de Sensibilidade Microbiana , Próteses e Implantes/microbiologia , Infecções Relacionadas à Prótese/microbiologia , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/isolamento & purificação , Vancomicina/farmacologia
7.
Int J Mol Sci ; 22(19)2021 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-34638591

RESUMO

Biofilm-related implant infections (BRII) are a disastrous complication of both elective and trauma orthopaedic surgery and occur when an implant becomes colonised by bacteria. The definitive treatment to eradicate the infections once a biofilm has established is surgical excision of the implant and thorough local debridement, but this carries a significant socioeconomic cost, the outcomes for the patient are often poor, and there is a significant risk of recurrence. Due to the large volumes of surgical procedures performed annually involving medical device implantation, both in orthopaedic surgery and healthcare in general, and with the incidence of implant-related infection being as high as 5%, interventions to prevent and treat BRII are a major focus of research. As such, innovation is progressing at a very fast pace; the aim of this study is to review the latest interventions for the prevention and treatment of BRII, with a particular focus on implant-related approaches.


Assuntos
Antibacterianos/uso terapêutico , Bactérias/efeitos dos fármacos , Biofilmes/efeitos dos fármacos , Procedimentos Ortopédicos/efeitos adversos , Complicações Pós-Operatórias/microbiologia , Próteses e Implantes/microbiologia , Animais , Humanos , Ortopedia/métodos
8.
Nat Commun ; 12(1): 5473, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34531396

RESUMO

Implant related infections are the most common cause of joint arthroplasty failure, requiring revision surgeries and a new implant, resulting in a cost of $8.6 billion annually. To address this problem, we created a class of coating technology that is applied in the operating room, in a procedure that takes less than 10 min, and can incorporate any desired antibiotic. Our coating technology uses an in situ coupling reaction of branched poly(ethylene glycol) and poly(allyl mercaptan) (PEG-PAM) polymers to generate an amphiphilic polymeric coating. We show in vivo efficacy in preventing implant infection in both post-arthroplasty infection and post-spinal surgery infection mouse models. Our technology displays efficacy with or without systemic antibiotics, the standard of care. Our coating technology is applied in a clinically relevant time frame, does not require modification of implant manufacturing process, and does not change the implant shelf life.


Assuntos
Antibacterianos/farmacologia , Materiais Revestidos Biocompatíveis/farmacologia , Sistemas Automatizados de Assistência Junto ao Leito , Infecções Relacionadas à Prótese/prevenção & controle , Infecções Estafilocócicas/prevenção & controle , Staphylococcus aureus/efeitos dos fármacos , Animais , Antibacterianos/química , Materiais Revestidos Biocompatíveis/química , Modelos Animais de Doenças , Humanos , Masculino , Camundongos Endogâmicos C57BL , Polietilenoglicóis/química , Polímeros/química , Próteses e Implantes/microbiologia , Próteses e Implantes/normas , Infecções Relacionadas à Prótese/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/fisiologia , Resultado do Tratamento
9.
NPJ Biofilms Microbiomes ; 7(1): 68, 2021 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-34385452

RESUMO

Hundreds of thousands of human implant procedures require surgical revision each year due to infection. Infections are difficult to treat with conventional antibiotics due to the formation of biofilm on the implant surface. We have developed a noninvasive method to eliminate biofilm on metal implants using heat generated by intermittent alternating magnetic fields (iAMF). Here, we demonstrate that heat and antibiotics are synergistic in biofilm elimination. For Pseudomonas aeruginosa biofilm, bacterial burden was reduced >3 log with iAMF and ciprofloxacin after 24 h compared with either treatment alone (p < 0.0001). This effect was not limited by pathogen or antibiotic as similar biofilm reductions were seen with iAMF and either linezolid or ceftriaxone in Staphylococcus aureus. iAMF and antibiotic efficacy was seen across various iAMF settings, including different iAMF target temperatures, dose durations, and dosing intervals. Initial mechanistic studies revealed membrane disruption as one factor important for AMF enhanced antibacterial activity in the biofilm setting. This study demonstrates the potential of utilizing a noninvasive approach to reduce biofilm off of metallic implants.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Biofilmes/efeitos da radiação , Campos Magnéticos , Metais , Bactérias/efeitos dos fármacos , Bactérias/efeitos da radiação , Ciprofloxacina/farmacologia , Farmacorresistência Bacteriana , Humanos , Testes de Sensibilidade Microbiana , Próteses e Implantes/microbiologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/efeitos da radiação , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/efeitos da radiação
10.
Methods Mol Biol ; 2341: 117-125, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34264467

RESUMO

Secreted bacterial proteins are difficult to identify directly from an infection site due to a limited amount of bacteria and presence of a large quantity of host proteins. Here we describe a rat model of orthopedic implant that allows us to harvest bacterial biofilm materials sufficient for identification of bacterial proteins in the biofilm matrix by liquid chromatography-tandem MS (GeLC-MS/MS) analysis.


Assuntos
Proteínas de Bactérias/isolamento & purificação , Biofilmes/crescimento & desenvolvimento , Infecções Relacionadas à Prótese/microbiologia , Infecções Estafilocócicas/diagnóstico , Staphylococcus aureus/fisiologia , Animais , Cromatografia Líquida , Modelos Animais de Doenças , Masculino , Próteses e Implantes/microbiologia , Ratos , Staphylococcus aureus/metabolismo , Espectrometria de Massas em Tandem
11.
Adv Sci (Weinh) ; 8(13): 2100681, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34258168

RESUMO

Implant infections caused by methicillin-resistant Staphylococcus aureus (MRSA) can cause major complications during the perioperative period. Diclofenac, one of the most widely used nonsteroidal anti-inflammatory drugs, is often used to relieve pain and inflammation. In this study, it is found that high-dose diclofenac can inhibit the growth of MRSA, and does not easily induce drug-resistant mutations after continuous passage. However, low-doses diclofenac can resensitize bacteria to ß-lactams, which help to circumvent drug resistance and improve the antibacterial efficacy of conventional antibiotics. Further, low-dose diclofenac in combination with ß-lactams inhibit MRSA associated biofilm formation in implants. Transcriptomic and proteomic analyses indicate that diclofenac can reduce the expression of genes and proteins associated with ß-lactam resistance: mecA, mecR, and blaZ; peptidoglycan biosynthesis: murA, murC, femA, and femB; and biofilm formation: altE and fnbP. Murine implant infection models indicate that diclofenac combined with ß-lactams, can substantially alleviate MRSA infections in vivo. In addition, it is investigated that low dose diclofenac can inhibit MRSA antibiotic resistance via the mecA/blaZ pathway and related biofilms in implants. The synergistic effect of diclofenac and ß-lactams might have promising applications for preventing perioperative infection, considering its multitarget effects against MRSA.


Assuntos
Antibacterianos/farmacologia , Anti-Inflamatórios não Esteroides/farmacologia , Diclofenaco/farmacologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Próteses e Implantes/microbiologia , beta-Lactamas/farmacologia , Animais , Modelos Animais de Doenças , Feminino , Camundongos
13.
Nat Commun ; 12(1): 3757, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34145249

RESUMO

Peptides are widely used for surface modification to develop improved implants, such as cell adhesion RGD peptide and antimicrobial peptide (AMP). However, it is a daunting challenge to identify an optimized condition with the two peptides showing their intended activities and the parameters for reaching such a condition. Herein, we develop a high-throughput strategy, preparing titanium (Ti) surfaces with a gradient in peptide density by click reaction as a platform, to screen the positions with desired functions. Such positions are corresponding to optimized molecular parameters (peptide densities/ratios) and associated preparation parameters (reaction times/reactant concentrations). These parameters are then extracted to prepare nongradient mono- and dual-peptide functionalized Ti surfaces with desired biocompatibility or/and antimicrobial activity in vitro and in vivo. We also demonstrate this strategy could be extended to other materials. Here, we show that the high-throughput versatile strategy holds great promise for rational design and preparation of functional biomaterial surfaces.


Assuntos
Materiais Revestidos Biocompatíveis/química , Próteses e Implantes/microbiologia , Titânio/química , Animais , Adesão Celular/fisiologia , Células Cultivadas , Ensaios de Triagem em Larga Escala , Camundongos , Coelhos , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Propriedades de Superfície
14.
Pathog Dis ; 79(6)2021 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-34124751

RESUMO

Orthopedic implant-associated bacterial infections with Staphylococcus aureus constitute a major clinical problem, and large pre-clinical animal models remain scarce. The aim of this study was to establish a standardized method of a localized, acute S. aureus bone infection in the presence of complex implanted devices in a sheep model. Four sheep underwent surgery receiving a complex implanted metallic device with a component stabilizing a bone defect created in the left tibial metaphysis, and an attached component placed in adjacent soft tissue. The bone defect was inoculated with S. aureus strain ATCC25293 (1 × 104 CFU). Twenty one days later, the surgery site was macroscopically evaluated, tissue samples and implants harvested for bacterial cell count quantification and tissue samples histologically analyzed. The animals exhibited clinical signs of localized infection (e.g. swelling, lameness, pain) but did not develop symptoms of sepsis. After euthanasia, macroscopic assessment revealed a localized bone and soft tissue infection at the surgery site. Histologically, an acute inflammation with neutrophils but also signs of bone destruction with necrosis was noted. An ovine model of a localized, acute S. aureus bone infection with complex implants was successfully established and could be used to test novel treatments against orthopedic implant-associated infections.


Assuntos
Osteomielite/microbiologia , Infecções Relacionadas à Prótese/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus , Animais , Biofilmes/crescimento & desenvolvimento , Modelos Animais de Doenças , Humanos , Osteomielite/diagnóstico por imagem , Osteomielite/patologia , Próteses e Implantes/microbiologia , Ovinos , Infecções Estafilocócicas/diagnóstico por imagem , Infecções Estafilocócicas/patologia
15.
ACS Appl Mater Interfaces ; 13(20): 23352-23368, 2021 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-33998809

RESUMO

Medical implant-associated infections resulting from biofilm formation triggered by unspecific protein adsorption are the prevailing cause of implant failure. However, implant surfaces rendered with multifunctional bioactive nanocoatings offer a promising alternative to prevent the initial attachment of bacteria and effectively interrupt biofilm formation. The need to research and develop novel and stable bioactive nanocoatings for medical implants and a comprehensive understanding of their properties in contact with the complex biological environment are crucial. In this study, we developed an aqueous stable and crosslinker-free polyelectrolyte-surfactant complex (PESC) composed of a renewable cationic polysaccharide, chitosan, a lysine-based anionic surfactant (77KS), and an amphoteric antibiotic, amoxicillin, which is widely used to treat a number of infections caused by bacteria. We successfully introduced the PESC as bioactive functional nanolayers on the "model" and "real" polydimethylsiloxane (PDMS) surfaces under dynamic and ambient conditions. Besides their high stability and improved wettability, these uniformly deposited nanolayers (thickness: 44-61 nm) with mixed charges exhibited strong repulsion toward three model blood proteins (serum albumin, fibrinogen, and γ-globulin) and their competitive interactions in the mixture in real-time, as demonstrated using a quartz crystal microbalance with dissipation (QCM-D). The functional nanolayers with a maximum negative zeta potential (ζ: -19 to -30 mV at pH 7.4), water content (1628-1810 ng cm-2), and hydration (low viscosity and elastic shear modulus) correlated with the mass, conformation, and interaction nature of proteins. In vitro antimicrobial activity testing under dynamic conditions showed that the charged nanolayers actively inhibited the growth of both Gram-negative (Escherichia coli) and Gram-positive (Staphylococcus aureus) bacteria compared to unmodified PDMS. Given the ease of fabrication of multifunctional and charged biobased coatings with simultaneous protein-repellent and antimicrobial activities, the limitations of individual approaches could be overcome leading to a better and advanced design of various medical devices (e.g., catheters, prosthetics, and stents).


Assuntos
Antibacterianos , Biofilmes/efeitos dos fármacos , Materiais Revestidos Biocompatíveis , Próteses e Implantes/microbiologia , Tensoativos , Antibacterianos/química , Antibacterianos/farmacologia , Quitosana/química , Quitosana/farmacologia , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Escherichia coli/efeitos dos fármacos , Interações Hidrofóbicas e Hidrofílicas , Lisina/química , Lisina/farmacologia , Nanomedicina , Nanoestruturas/química , Proteínas/química , Silício , Staphylococcus aureus/efeitos dos fármacos , Propriedades de Superfície , Tensoativos/química , Tensoativos/farmacologia
16.
Clin Orthop Relat Res ; 479(7): 1613-1623, 2021 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-33847603

RESUMO

BACKGROUND: We developed iodine-coated titanium implants to suppress microbial activity and prevent periprosthetic joint infection (PJI); their efficacy was demonstrated in animal and in vitro models. The iodine content in iodine-coated implants naturally decreases in vivo. However, to our knowledge, the effect of reduced iodine content on the implant's antimicrobial activity has not been evaluated to date. QUESTIONS/PURPOSES: (1) How much does the iodine content on the implant surface decrease after 4 and 8 weeks in vivo in a rat model? (2) What effect does the reduced iodine content have on the antimicrobial effect of the implant against multiple bacteria in an in vitro model? METHODS: This experiment was performed in two parts: an in vivo experiment to determine attenuation of iodine levels over time in rats, and an in vitro experiment in which we sought to assess whether the reduced iodine content observed in the in vivo experiment was still sufficient to deliver antimicrobial activity against common pathogens seen in PJI. For the in vivo experiment, three types of titanium alloy washers were implanted in rats: untreated (Ti), surface-anodized to produce an oxide film (Ti-O), and with an iodine layer on the oxidation film (Ti-I). The attenuation of iodine levels in rats was measured over time using inductively coupled plasma-mass spectrometry. Herein, only the Ti-I washer was used, with five implanted in each rat that were removed after 4 or 8 weeks. For the 4- and 8-week models, two rats and 15 washers were used. For the in vitro study, to determine the antibacterial effect, three types of washers (Ti, Ti-O, and Ti-I) (nine washers in total) were implanted in each rat. Then, the washers were removed and the antibacterial effect of each washer was examined on multiple bacterial species using the spread plate method and fluorescence microscopy. For the spread plate method, six rats were used, and five rats were used for the observation using fluorescence microscopy; further, 4- and 8-week models were made for each method. Thus, a total of 22 rats and 198 washers were used. Live and dead bacteria in the biofilm were stained, and the biofilm coverage percentage for quantitative analysis was determined using fluorescence microscopy in a nonblinded manner. Ti-I was used as the experimental group, and Ti and Ti-O were used as control groups. The total number of rats and washers used throughout this study was 24 and 213, respectively. RESULTS: Iodine content in rats implanted with Ti-I samples decreased to 72% and 65% after the in vivo period of 4 and 8 weeks, respectively (p = 0.001 and p < 0.001, respectively). In the in vitro experiment, the Ti-I implants demonstrated a stronger antimicrobial activity than Ti and Ti-O implants in the 4- and 8-week models. Both the median number of bacterial colonies and the median biofilm coverage percentage with live bacteria on Ti-I were lower than those on Ti or Ti-O implants for each bacterial species in the 4- and 8-week models. There was no difference in the median biofilm coverage percentage of dead bacteria. In the 8-week model, the antibacterial activity using the spread plate method had median (interquartile range) numbers of bacteria on the Ti, Ti-O, and Ti-I implants of 112 (104 to 165) × 105, 147 (111 to 162) × 105, and 55 (37 to 67) × 105 of methicillin-sensitive Staphylococcus aureus (Ti-I versus Ti, p = 0.026; Ti-I versus Ti-O, p = 0.009); 71 (39 to 111) × 105, 50 (44 to 62) × 105, and 26 (9 to 31)× 105 CFU of methicillin-resistant S. aureus (Ti-I versus Ti, p = 0.026; Ti-I versus Ti-O, p = 0.034); and 77 (74 to 83) × 106, 111 (95 to 117) × 106, and 30 (21 to 45) × 106 CFU of Pseudomonas aeruginosa (Ti-I versus Ti, p = 0.004; Ti-I versus Ti-O, p = 0.009). Despite the decrease in the iodine content of Ti-I after 8 weeks, it demonstrated better antibacterial activity against all tested bacteria than the Ti and Ti-O implants. CONCLUSION: Iodine-coated implants retained their iodine content and antibacterial activity against methicillin-sensitive S. aureus, methicillin-resistant S. aureus, and P. aeruginosa for 8 weeks in vivo in rats. To evaluate the longer-lasting antibacterial efficacy, further research using larger infected animal PJI models with implants in the joints of both males and females is desirable. CLINICAL RELEVANCE: Iodine-coated titanium implants displayed an antibacterial activity for 8 weeks in rats in vivo. Although the findings in a rat model do not guarantee efficacy in humans, they represent an important step toward clinical application.


Assuntos
Antibacterianos/farmacologia , Biofilmes/efeitos dos fármacos , Materiais Revestidos Biocompatíveis/farmacologia , Iodo/farmacologia , Infecções Relacionadas à Prótese/prevenção & controle , Animais , Modelos Animais de Doenças , Humanos , Técnicas In Vitro , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Próteses e Implantes/microbiologia , Desenho de Prótese , Infecções Relacionadas à Prótese/microbiologia , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/prevenção & controle , Pseudomonas aeruginosa/efeitos dos fármacos , Ratos , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/prevenção & controle , Titânio
17.
Int J Infect Dis ; 106: 302-307, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33819605

RESUMO

OBJECTIVES: In the microbiological diagnosis of periprosthetic joint infection (PJI), there is much discussion about the methodology of obtaining proper specimens, the processing technique, and suitable culture media. This retrospective study was conducted to analyse the accuracy of our culture techniques. METHODS: Tissue samples and components from 258 patients after revision arthroplasty of the hip, knee, and shoulder were investigated, and the results of tissue cultures (TC) were compared to those of sonicate fluid cultures (SFC). Furthermore, an evaluation was performed of the influence of different culture media on the detection rate. RESULTS: PJI was confirmed in 186 patients. The overall sensitivity of TC was no different to that of SFC (91.3% vs 90.8%, P = 1). In 153 cases (82.3%), TC and SFC showed concordant positive results. Results were discordant in 33 cases (17.7%). When differentiated according to the type of infection, TC showed significantly better results than SFC in detecting polymicrobial infections (97.0% vs 67.0%, P = 0.004). There were also significant differences between the culture media regarding the yield of microorganisms. CONCLUSIONS: TC was more effective in detecting co-infections. The best results were obtained using both TC and SFC. The choice of culture media has a significant influence on the quality of results.


Assuntos
Artrite Infecciosa/diagnóstico , Próteses e Implantes/microbiologia , Infecções Relacionadas à Prótese/diagnóstico , Sonicação , Adulto , Idoso , Idoso de 80 Anos ou mais , Artrite Infecciosa/patologia , Artroplastia/efeitos adversos , Humanos , Masculino , Pessoa de Meia-Idade , Infecções Relacionadas à Prótese/microbiologia , Infecções Relacionadas à Prótese/patologia , Estudos Retrospectivos
18.
Acta Biochim Biophys Sin (Shanghai) ; 53(6): 683-696, 2021 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-33772282

RESUMO

The bacteria-mediated inflammatory conditions adversely affect the osseointegration process of endosseous implants, which can even lead to implant malfunction or failure. Local drug delivery has been designed to exert anti-inflammatory and antibacterial activities, but whether this strategy has an effect on the compromised osseointegration under inflammation has rarely been studied. The present study focused on the osteoinductive efficacy of two known phytoestrogens [bergapten (BP) and quercetin (QE)] on implant sites under multiple bacteria-infected conditions in situ. Furthermore, the gene expression profiles of rat bone mesenchymal stem cells (rBMSCs) treated with BP and QE in the presence of Porphyromonas gingivalis-derived lipopolysaccharide were identified. The results showed that both drugs, especially QE, had significant potentiating effects on promoting osteogenic differentiation of rBMSCs, resisting multiple pathogens, and reducing inflammatory activity. Meanwhile, RNA sequencing analysis highlighted the enriched gene ontology terms and the differentially expressed genes (Vps25, Il1r2, Csf3, Efemp1, and Ccl20) that might play essential roles in regulating the above tri-effects, which provided the basis for the drug delivery system to be used as a novel therapeutic strategy for integrating peri-implant health. Overall, our study confirmed that QE appeared to outperform BP in osteogenesis and bacterial killing but not in anti-inflammation. Moreover, both drugs possess favorable tri-effects and can serve as the pivotal agents for the drug delivery system to boost osseointegration at inflammatory implant sites.


Assuntos
5-Metoxipsoraleno/farmacologia , Antibacterianos/farmacologia , Anti-Inflamatórios/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteogênese/efeitos dos fármacos , Fitoestrógenos/farmacologia , Quercetina/farmacologia , Animais , Biofilmes/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Sistemas de Liberação de Medicamentos/métodos , Lipopolissacarídeos/farmacologia , Masculino , Osseointegração/efeitos dos fármacos , Porphyromonas gingivalis/fisiologia , Próteses e Implantes/microbiologia , Ratos , Ratos Sprague-Dawley , Transcriptoma/efeitos dos fármacos
19.
J Biomater Appl ; 36(3): 441-459, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33599133

RESUMO

The bacterial contamination in implants has been recognized as one of the key issues in orthopedics. In this article, a new technique of electrical polarization of various non-piezoelectric and piezoelectric biocompatible ceramics has been explored to develop antibacterial implants. Optimally processed hydroxyapatite (HA), BaTiO3 (BT), CaTiO3 (CT), Na0.5K0.5NbO3 (NKN) and their composites have been used as model biomaterials to verify the concept. The phase evolution analyses and microstructural characterizations were performed for sintered samples. The samples were polarized at polarizing voltage and temperature of 20 kV and 500°C, respectively, for 30 min. The hydrophilicity of polarized surfaces was examined using deionized water and culture media. The polarization induced in-vitro antibacterial study was performed for both, gram positive and gram negative bacteria. The viability of Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) bacteria reduces significantly on the polarized surfaces. In addition, the influence of polarization on antibacterial response has been explored via various mechanisms such as development of reactive oxygen species (ROS), catalase activity and lipoperoxidation. Furthermore, the cellular response of polarized surfaces was also examined using SaOS2 and MG-63 cells. The viability of SaOS2 and MG-63 cells was observed to increase significantly on negatively polarized surfaces. Overall, the surface treatment enhances the antibacterial response of HA, NKN, BT, CT and their composites surfaces with positive influence on cellular response.


Assuntos
Antibacterianos/química , Materiais Biocompatíveis/química , Cerâmica/química , Antibacterianos/farmacologia , Materiais Biocompatíveis/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cerâmica/farmacologia , Durapatita/química , Durapatita/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Humanos , Próteses e Implantes/microbiologia , Eletricidade Estática
20.
J Appl Microbiol ; 131(3): 1498-1514, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33565669

RESUMO

AIMS: Joint infections cause premature implant failure. The avoidance of bacterial colonization of implant materials by modification of the material surface is therefore the focus of current research. In this in vitro study the complex interaction of periodic structures on PET and titanium surfaces on the adhesion of Staphylococcus aureus is analysed. METHODS AND RESULTS: Using direct laser interference patterning as well as roll-to-roll hot embossing methods, structured periodic textures of different spatial distance were produced on surfaces and S. aureus were cultured for 24 h on these. The amount of adhering bacteria was quantified using fluorescence microscopy and the local adhesion behaviour was investigated using scanning electron microscopy. For PET structures, minimal bacterial adhesion was identified for an aspect ratio of about 0·02. On titanium structures, S. aureus adhesion was significantly decreased for profile heights of < 200 nm. Our results show a significantly decreased bacterial adhesion for structures with an aspect ratio range of 0·02 to 0·05. CONCLUSIONS: We show that structuring on surfaces can decrease the amount of S. aureus on titanium and PET as common implant materials. SIGNIFICANCE AND IMPACT OF THE STUDY: The study highlights the immense potential of applying specific structures to implant materials to prevent implant colonization with pathogen bacteria.


Assuntos
Aderência Bacteriana , Polietilenotereftalatos/química , Staphylococcus aureus , Titânio/química , Próteses e Implantes/microbiologia , Staphylococcus aureus/fisiologia , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...