Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
EFORT Open Rev ; 9(5): 339-348, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38726986

RESUMO

The treatment of musculoskeletal infections (MSIs), including periprosthetic joint infection (PJI) and fracture-related infection (FRI), is often complicated by biofilm-related challenges necessitating multiple revision surgeries and incurring substantial costs. The emergence of antimicrobial resistance (AMR) adds to the complexity of the problem, leading to increased morbidity and healthcare expenses. There is an urgent need for novel antibacterial strategies, with the World Health Organization endorsing non-traditional approaches like bacteriophage (phage) therapy. Phage therapy, involving the targeted application of lytic potent phages, shows promise in the treatment of MSIs. Although historical clinical trials and recent case studies present significant milestones in the evolution of phage therapy over the past century, challenges persist, including variability in study designs, administration protocols and phage selection. Efforts to enhance treatment efficacy consist of personalized phage therapy and combination with antibiotics. Future perspectives entail addressing regulatory barriers, standardizing treatment protocols, and conducting high-quality clinical trials to establish phage therapy's efficacy for the treatment of MSIs. Initiatives like the PHAGEFORCE study and the PHAGEinLYON Clinic programme aim to streamline phage therapy, facilitating personalized treatment approaches and systematic data collection to advance its clinical utility in these challenging infections.

2.
ACS Nano ; 18(11): 8017-8028, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38456817

RESUMO

d-Amino acids are signals for biofilm disassembly. However, unexpected metabolic pathways severely attenuate the utilization of d-amino acids in biofilm disassembly, resulting in unsatisfactory efficiency. Herein, three-dimensional poly(d-amino acid) nanoparticles (NPs), which possess the ability to block intracellular metabolism, are constructed with the aim of disassembling the biofilms. The obtained poly(α-N-acryloyl-d-phenylalanine)-block-poly(ß-N-acryloyl-d-aminoalanine NPs (denoted as FA NPs) present α-amino groups and α-carboxyl groups of d-aminoalanine on their surface, which guarantees that FA NPs can effectively insert into bacterial peptidoglycan (PG) via the mediation of PG binding protein 4 (PBP4). Subsequently, the FA NPs trigger the detachment of amyloid-like fibers that connect to the PG and reduce the number of polysaccharides and proteins in extracellular polymeric substances (EPS). Finally, FA NPs damage the structural stability of EPS and lead to the disassembly of the biofilm. Based on this feature, FA NPs significantly enhance the killing efficacy of encapsulated sitafloxacin sesquihydrate (Sita) by facilitating the penetration of Sita within the biofilm, achieving complete elimination of Staphylococcal biofilm in mice. Therefore, this study strongly demonstrates that FA NPs can effectively improve biofilm disassembly efficacy and provide great potential for bacterial biofilm infection treatment.


Assuntos
Aminoácidos , Nanopartículas , Animais , Camundongos , Aminoácidos/química , Peptidoglicano , Biofilmes , Polissacarídeos , Nanopartículas/química
3.
Clin Microbiol Infect ; 29(6): 695-701, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36669559

RESUMO

BACKGROUND: Bacteriophage therapy has a long history in the treatment of musculoskeletal and skin/soft tissue infections, particularly in the former Soviet Union. Due to the global rise in antimicrobial resistance, phage application has experienced a resurgence of interest and expanded to many countries. OBJECTIVES: This narrative review aims to provide clinical microbiologists, infectious disease specialists and surgeons a brief history of bacteriophage therapy for human musculoskeletal and soft tissue infections, as well as data on current practices and ongoing clinical studies. SOURCES: A search of PubMed and Clinicaltrials.gov was performed to identify relevant studies. Search terms were 'bacteriophage therapy', 'musculoskeletal infection' and 'soft tissue infection'. The bibliography of all retrieved articles was checked for additional relevant references. CONTENT: Past and current data on the use of bacteriophage therapy for human musculoskeletal, skin and soft tissue infections are evaluated. Moreover, we present the clinical trials registered in public databases. Based on current clinical experience and data, several scenarios of bacteriophage application for human therapy are examined. Finally, we discuss legislative hurdles in the regulatory approval process and present future perspectives for bacteriophage therapy. IMPLICATIONS: Antimicrobial resistance is one of the most important global public health challenges. Several different alternatives to conventional antibiotics are under development; bacteriophage therapy is one of them. Currently, therapeutic use of phages is restrained by regulatory hurdles and largely limited to sporadic authorization in compassionate use or under temporary approval as new drugs in Europe and the US. Although bacteriophage therapy seems to be safe and clinical results of phage treatment are promising, future data from high-quality (randomized controlled) trials could provide a better understanding of the reasonable minimal criteria required for expansion of bacteriophage therapy.


Assuntos
Bacteriófagos , Terapia por Fagos , Infecções dos Tecidos Moles , Humanos , Infecções dos Tecidos Moles/tratamento farmacológico , Antibacterianos/uso terapêutico , Terapia por Fagos/métodos , Europa (Continente)
4.
Indian J Orthop ; 57(1): 44-51, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36660477

RESUMO

Background: Musculoskeletal infections are a major source of morbidity for orthopedic and trauma patients, are associated with prolonged treatment times, and, unfortunately, suffer from poor functional outcomes. Further complicating the issue, antimicrobial resistance (AMR) is increasingly impacting the treatment of musculoskeletal infections with a diminishing repertoire of effective antibiotic agents for some highly resistant pathogens. Most orthopedic surgical procedures involve implants, and the formation of bacterial biofilms on these implants is now recognized as a major factor contributing to the failure of antibiotic therapy in orthopedic surgery. Methods: This review presents an overview of the types, structure, formation, and pathogenesis of biofilms as they pertain to musculoskeletal infections. Furthermore, it describes the key concepts in the management of biofilms and future perspectives for the better treatment of patients with biofilm-related musculoskeletal infections. Results: A bacterial biofilm is a dynamic, living conglomerate of bacteria encased in an extracapsular polysaccharide matrix (EPS). Biofilms are a natural mode of survival for virtually all bacterial species, including both Grampositive and Gram-negative bacteria, as well as fungi. The biofilm model of growth confers resistance by several well-defined mechanisms regardless of the species of the microorganism. In most cases, biofilm management often necessitates radical measures to ensure eradication including both surgical and medical interventions. Conclusions: Orthopedic surgeons should be aware of the key concepts pertaining to biofilms, and the impact that these can have on clinical practice.

5.
Macromol Biosci ; 23(1): e2200311, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36189899

RESUMO

The intracellular survival of pathogenic bacteria requires a range of survival strategies and virulence factors. These infections are a significant clinical challenge, wherein treatment frequently fails because of poor antibiotic penetration, stability, and retention in host cells. Drug delivery systems (DDSs) are promising tools to overcome these shortcomings and enhance the efficacy of antibiotic therapy. In this review, the classification and the mechanisms of intracellular bacterial persistence are elaborated. Furthermore, the systematic design strategies applied to DDSs to eliminate intracellular bacteria are also described, and the strategies used for internalization, intracellular activation, bacterial targeting, and immune enhancement are highlighted. Finally, this overview provides guidance for constructing functionalized DDSs to effectively eliminate intracellular bacteria.


Assuntos
Nanopartículas , Sistemas de Liberação de Medicamentos , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Bactérias
6.
Front Cell Infect Microbiol ; 12: 910970, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35811672

RESUMO

S. aureus infection of bone is difficult to eradicate due to its ability to colonize the osteocyte-lacuno-canalicular network (OLCN), rendering it resistant to standard-of-care (SOC) antibiotics. To overcome this, we proposed two bone-targeted bisphosphonate-conjugated antibiotics (BCA): bisphosphonate-conjugated sitafloxacin (BCS) and hydroxybisphosphonate-conjugate sitafloxacin (HBCS). Initial studies demonstrated that the BCA kills S. aureus in vitro. Here we demonstrate the in vivo efficacy of BCS and HBCS versus bisphosphonate, sitafloxacin, and vancomycin in mice with implant-associated osteomyelitis. Longitudinal bioluminescent imaging (BLI) confirmed the hypothesized "target and release"-type kinetics of BCS and HBCS. Micro-CT of the infected tibiae demonstrated that HBCS significantly inhibited peri-implant osteolysis versus placebo and free sitafloxacin (p < 0.05), which was not seen with the corresponding non-antibiotic-conjugated bisphosphonate control. TRAP-stained histology confirmed that HBCS significantly reduced peri-implant osteoclast numbers versus placebo and free sitafloxacin controls (p < 0.05). To confirm S. aureus killing, we compared the morphology of S. aureus autolysis within in vitro biofilm and infected tibiae via transmission electron microscopy (TEM). Live bacteria in vitro and in vivo presented as dense cocci ~1 µm in diameter. In vitro evidence of autolysis presented remnant cell walls of dead bacteria or "ghosts" and degenerating (non-dense) bacteria. These features of autolyzed bacteria were also present among the colonizing S. aureus within OLCN of infected tibiae from placebo-, vancomycin-, and sitafloxacin-treated mice, similar to placebo. However, most of the bacteria within OLCN of infected tibiae from BCA-treated mice were less dense and contained small vacuoles and holes >100 nm. Histomorphometry of the bacteria within the OLCN demonstrated that BCA significantly increased their diameter versus placebo and free antibiotic controls (p < 0.05). As these abnormal features are consistent with antibiotic-induced vacuolization, bacterial swelling, and necrotic phenotype, we interpret these findings to be the initial evidence of BCA-induced killing of S. aureus within the OLCN of infected bone. Collectively, these results support the bone targeting strategy of BCA to overcome the biodistribution limits of SOC antibiotics and warrant future studies to confirm the novel TEM phenotypes of bacteria within OLCN of S. aureus-infected bone of animals treated with BCS and HBCS.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Osteomielite , Infecções Estafilocócicas , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Difosfonatos/farmacologia , Difosfonatos/uso terapêutico , Modelos Animais de Doenças , Fluoroquinolonas , Camundongos , Osteomielite/tratamento farmacológico , Osteomielite/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus , Distribuição Tecidual , Vancomicina/farmacologia
7.
Front Cell Infect Microbiol ; 12: 826392, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35573772

RESUMO

Local antibiotic therapy is increasingly being recognised for its role in preventing and treating orthopaedic device-related infection (ODRI). A bioresorbable, injectable gentamicin-loaded hydrogel has been developed to deliver local antibiotics at the time of surgery with potential for both prevention and treatment of ODRI. In a prophylaxis model, the antibiotic hydrogel was compared with systemic perioperative antibiotic prophylaxis alone in twelve sheep (six per group) at the time of intramedullary (IM) nail insertion to the tibia, which was inoculated with methicillin-sensitive Staphylococcus aureus (MSSA). In a treatment model of single-stage revision surgery, adjunctive antibiotic-loaded hydrogel was compared with systemic antibiotics alone in a single stage revision of MSSA infection associated with a tibia intramedullary nail in eleven sheep (five/six per group). The primary endpoint was quantitative microbiological results of soft tissue, bone and sonicate fluid from explanted hardware at the time of euthanasia. At euthanasia, the control sheep that received no local antibiotics in the prophylaxis model were all culture-positive (median 1x108, range 7x106-3x108 colony forming units, CFU) while only two of six sheep receiving local gentamicin had any culture positive biopsies (median 1x101, range 0 - 1x105 CFU). For the treatment model, sheep receiving only systemic antibiotics were all culture-positive (median 8x105, range 2x103- 9x106 CFU) while only two of six sheep treated with gentamicin-loaded hydrogel had any culture positive biopsies (median 3x102, range 0 - 7x104 CFU). Local gentamicin concentrations measured in extracellular fluid in the tibial canal show a burst release of gentamicin from the hydrogel. Serum gentamicin concentrations peaked in both models at one day post application and were below detection limit thereafter. This study has demonstrated the effective use of a locally delivered antibiotic hydrogel for both the prevention and treatment of ODRI that is superior to that of systemic antibiotics alone. Future studies will endeavour to translate from preclinical to clinical research trials.


Assuntos
Ortopedia , Infecções Estafilocócicas , Animais , Antibacterianos/uso terapêutico , Modelos Animais de Doenças , Gentamicinas , Hidrogéis , Ovinos , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/prevenção & controle
9.
Bone Joint Res ; 10(1): 77-84, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33474969

RESUMO

AIMS: Biofilm formation is one of the primary reasons for the difficulty in treating implant-related infections (IRIs). Focused high-energy extracorporeal shockwave therapy (fhESWT), which is a treatment modality for fracture nonunions, has been shown to have a direct antibacterial effect on planktonic bacteria. The goal of the present study was to investigate the effect of fhESWT on Staphylococcus aureus biofilms in vitro in the presence and absence of antibiotic agents. METHODS: S. aureus biofilms were grown on titanium discs (13 mm × 4 mm) in a bioreactor for 48 hours. Shockwaves were applied with either 250, 500, or 1,000 impulses onto the discs surrounded by either phosphate-buffered saline or antibiotic (rifampin alone or in combination with nafcillin). The number of viable bacteria was determined by quantitative culture after sonication. Representative samples were taken for scanning electron microscopy. RESULTS: The application of fhESWT led to a ten-fold reduction in bacterial counts on the metal discs for all impulse numbers compared to the control (p < 0.001). Increasing the number of impulses did not further reduce bacterial counts in the absence of antibiotics (all p > 0.289). Antibiotics alone reduced the number of bacteria on the discs; however, the combined application of the fhESWT and antibiotic administration further reduced the bacterial count compared to the antibiotic treatment only (p = 0.032). CONCLUSION: The use of fhESWT significantly reduced the colony-forming unit (CFU) count of a S. aureus biofilm in our model independently, and in combination with antibiotics. Therefore, the supplementary application of fhESWT could be a helpful tool in the treatment of IFIs in certain cases, including infected nonunions. Cite this article: Bone Joint Res 2021;10(1):77-84.

10.
ACS Appl Bio Mater ; 4(8): 6219-6226, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35006889

RESUMO

A bacterial biofilm is one of the main reasons that many diseases are difficult to cure. Herein, a teicoplanin (TPN)-loaded self-adapting chitosan-based hydrogel (CPH) system, called TPN-CPH, was prepared by encapsulating antibacterial TPN into CPH. This TPN-CPH can effectively combat preformed biofilms in vitro of Staphylococcus aureus (S. aureus). It has a good therapeutic effect on full-thickness cutaneous wounds in vivo of mice infected with biofilms. In addition, TPN-CPH can accelerate wound healing by self-adapting the wound and providing a moist environment. The operation process of TPN-CPH is simple, and no external stimulation such as light and heat is needed in the treatment process, making it more convenient for clinical application. Furthermore, this is a challenge to use self-adapting hydrogels to adapt the micro-size channels of biofilms. TPN-CPH provides a chitosan-based self-adapting hydrogel system for loading drugs to kill bacteria in biofilms, and thus it is promising for infection control.


Assuntos
Quitosana , Hidrogéis , Animais , Bactérias , Biofilmes , Quitosana/farmacologia , Hidrogéis/farmacologia , Camundongos , Staphylococcus aureus , Teicoplanina/farmacologia , Cicatrização
11.
J Orthop Res ; 39(2): 281-290, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32735351

RESUMO

Fracture-related infection (FRI) is a serious complication following musculoskeletal trauma. Accurate diagnosis and appropriate treatment depend on retrieving adequate deep tissue biopsies for bacterial culture. The aim of this cohort study was to compare intraoperative tissue cultures obtained by the Reamer-Irrigator-Aspirator system (RIA)-system against standard tissue cultures obtained during the same surgical procedure. All patients had long bone fractures of the lower limbs and were assigned to the FRI or Control group based on the FRI consensus definition. The FRI group consisted of 24 patients with confirmed FRI and the Control group consisted of 21 patients with aseptic nonunion or chronic pain (in the absence of other suggestive/confirmatory criteria). Standard tissue cultures and cultures harvested by the RIA-system showed similar results. In the FRI group, standard tissue cultures and RIA cultures revealed relevant pathogens in 67% and 71% of patients, respectively. Furthermore, in four FRI patients, cultures obtained by the RIA-system revealed additional relevant pathogens that were not found by standard tissue culturing, which contributed to the optimization of the treatment plan. In the Control group, there were no false-positive RIA culture results. As a proof-of-concept, this cohort study showed that the RIA-system could have a role in the diagnosis of FRI as an adjunct to standard tissue cultures. Since scientific evidence on the added value of the RIA-system in the management of FRI is currently limited, further research on this topic is required before its routine application in clinical practice.


Assuntos
Fraturas Ósseas/complicações , Procedimentos Ortopédicos/instrumentação , Infecção da Ferida Cirúrgica/microbiologia , Adulto , Feminino , Fraturas Ósseas/cirurgia , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Infecção da Ferida Cirúrgica/diagnóstico , Infecção da Ferida Cirúrgica/cirurgia , Técnicas de Cultura de Tecidos
12.
Front Microbiol ; 11: 538060, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33072008

RESUMO

As viruses with high specificity for their bacterial hosts, bacteriophages (phages) are an attractive means to eradicate bacteria, and their potential has been recognized by a broad range of industries. Against a background of increasing rates of antibiotic resistance in pathogenic bacteria, bacteriophages have received much attention as a possible "last-resort" strategy to treat infections. The use of bacteriophages in human patients is limited by their sensitivity to acidic pH, enzymatic attack and short serum half-life. Loading phage within a biomaterial can shield the incorporated phage against many of these harmful environmental factors, and in addition, provide controlled release for prolonged therapeutic activity. In this review, we assess the different classes of biomaterials (i.e., biopolymers, synthetic polymers, and ceramics) that have been used for phage delivery and describe the processing methodologies that are compatible with phage embedding or encapsulation. We also elaborate on the clinical or pre-clinical data generated using these materials. While a primary focus is placed on the application of phage-loaded materials for treatment of infection, we also include studies from other translatable fields such as food preservation and animal husbandry. Finally, we summarize trends in the literature and identify current barriers that currently prevent clinical application of phage-loaded biomaterials.

13.
J Biomed Mater Res B Appl Biomater ; 107(4): 1095-1106, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30332531

RESUMO

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.


Assuntos
Doenças Ósseas Infecciosas , Implantes Experimentais/microbiologia , Cetonas/química , Teste de Materiais , Polietilenoglicóis/química , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Animais , Benzofenonas , Doenças Ósseas Infecciosas/imunologia , Doenças Ósseas Infecciosas/microbiologia , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Osseointegração , Polímeros , Infecções Estafilocócicas/patologia
14.
J Orthop Trauma ; 32(4): 190-195, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29558373

RESUMO

OBJECTIVES: The optimal duration of perioperative antibiotic prophylaxis (PAP) for open fractures remains controversial because of heterogeneous or unclear guidelines and highly variable prophylactic regimens in clinical practice. We aimed at testing different PAP durations under controlled conditions in a contaminated rabbit fracture model. METHODS: A complete humeral osteotomy in 18 rabbits was fixed with a 7-hole locking compression plate and inoculated with Staphylococcus aureus. Cefuroxime was administered in a weight-adjusted dosage equivalent to human medicine (18.75 mg/kg). PAP was administered as a single shot only; for 24 hours; or for 72 hours in separate groups of rabbits (n = 6 per group). Infection was assessed after 2 weeks by quantitative bacteriological evaluation of the tissues and hardware. RESULTS: Postoperative duration of PAP had a significant impact on the success of antibiotic prophylaxis in this model. Whereas the single-shot regimen completely failed to prevent infection, the 24-hour regimen showed a reduced infection rate (1 of 6 rabbits infected), but only the 72-hour course was able to prevent fracture-related infection in all animals in our model. CONCLUSIONS: When contamination with high bacterial loads is likely (eg, in an open fracture situation), a 72-hour course of intravenous cefuroxime seems to be superior in preventing fracture-related infection in our rabbit model compared with a single-shot or 24-hour antibiotic regimen.


Assuntos
Antibacterianos/uso terapêutico , Antibioticoprofilaxia , Cefuroxima/uso terapêutico , Fixação Interna de Fraturas/efeitos adversos , Fraturas Expostas/complicações , Fraturas do Úmero/complicações , Infecções Estafilocócicas/prevenção & controle , Animais , Placas Ósseas , Modelos Animais de Doenças , Fraturas Expostas/cirurgia , Fraturas do Úmero/cirurgia , Coelhos , Infecções Estafilocócicas/etiologia , Staphylococcus aureus
16.
Appl Environ Microbiol ; 82(24): 7185-7196, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27736791

RESUMO

The immune-modulating properties of certain bifidobacterial strains, such as Bifidobacterium longum subsp. longum 35624 (B. longum 35624), have been well described, although the strain-specific molecular characteristics associated with such immune-regulatory activity are not well defined. It has previously been demonstrated that B. longum 35624 produces a cell surface exopolysaccharide (sEPS), and in this study, we investigated the role played by this exopolysaccharide in influencing the host immune response. B. longum 35624 induced relatively low levels of cytokine secretion from human dendritic cells, whereas an isogenic exopolysaccharide-negative mutant derivative (termed sEPSneg) induced vastly more cytokines, including interleukin-17 (IL-17), and this response was reversed when exopolysaccharide production was restored in sEPSneg by genetic complementation. Administration of B. longum 35624 to mice of the T cell transfer colitis model prevented disease symptoms, whereas sEPSneg did not protect against the development of colitis, with associated enhanced recruitment of IL-17+ lymphocytes to the gut. Moreover, intranasal administration of sEPSneg also resulted in enhanced recruitment of IL-17+ lymphocytes to the murine lung. These data demonstrate that the particular exopolysaccharide produced by B. longum 35624 plays an essential role in dampening proinflammatory host responses to the strain and that loss of exopolysaccharide production results in the induction of local TH17 responses. IMPORTANCE: Particular gut commensals, such as B. longum 35624, are known to contribute positively to the development of mucosal immune cells, resulting in protection from inflammatory diseases. However, the molecular basis and mechanisms for these commensal-host interactions are poorly described. In this report, an exopolysaccharide was shown to be decisive in influencing the immune response to the bacterium. We generated an isogenic mutant unable to produce exopolysaccharide and observed that this mutation caused a dramatic change in the response of human immune cells in vitro In addition, the use of mouse models confirmed that lack of exopolysaccharide production induces inflammatory responses to the bacterium. These results implicate the surface-associated exopolysaccharide of the B. longum 35624 cell envelope in the prevention of aberrant inflammatory responses.


Assuntos
Infecções por Bifidobacteriales/imunologia , Bifidobacterium longum/imunologia , Polissacarídeos Bacterianos/imunologia , Células Th17/imunologia , Animais , Infecções por Bifidobacteriales/microbiologia , Bifidobacterium longum/genética , Citocinas/imunologia , Feminino , Humanos , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos BALB C
17.
J Clin Microbiol ; 52(5): 1595-606, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24599975

RESUMO

Propionibacterium acnes and coagulase-negative staphylococci (CoNS) are opportunistic pathogens implicated in prosthetic joint and fracture fixation device-related infections. The purpose of this study was to determine whether P. acnes and the CoNS species Staphylococcus lugdunensis, isolated from an "aseptically failed" prosthetic hip joint and a united intramedullary nail-fixed tibial fracture, respectively, could cause osteomyelitis in an established implant-related osteomyelitis model in rabbits in the absence of wear debris from the implant material. The histological features of P. acnes infection in the in vivo rabbit model were consistent with localized pyogenic osteomyelitis, and a biofilm was present on all explanted intramedullary (IM) nails. The animals displayed no outward signs of infection, such as swelling, lameness, weight loss, or elevated white blood cell count. In contrast, infection with S. lugdunensis resulted in histological features consistent with both pyogenic osteomyelitis and septic arthritis, and all S. lugdunensis-infected animals displayed weight loss and an elevated white blood cell count despite biofilm detection in only two out of six rabbits. The differences in the histological and bacteriological profiles of the two species in this rabbit model of infection are reflective of their different clinical presentations: low-grade infection in the case of P. acnes and acute infection for S. lugdunensis. These results are especially important in light of the growing recognition of chronic P. acnes biofilm infections in prosthetic joint failure and nonunion of fracture fixations, which may be currently reported as "aseptic" failure.


Assuntos
Fixação Intramedular de Fraturas/efeitos adversos , Osteomielite/microbiologia , Propionibacterium acnes/isolamento & purificação , Staphylococcus lugdunensis/isolamento & purificação , Animais , Artrite Infecciosa/microbiologia , Biofilmes/crescimento & desenvolvimento , Feminino , Articulação do Quadril/microbiologia , Humanos , Coelhos , Infecções Estafilocócicas/microbiologia , Tíbia/microbiologia
18.
J Orthop Trauma ; 25(8): 488-93, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21738062

RESUMO

Preclinical modeling of human disease with animals has not been standardized for many common pathologic processes. Assorted animal models are being used to investigate the pathogenesis, prevention, and treatment of disease processes. Certainly it is difficult to interpret the current literature because there are diverse and often irrelevant models being implemented. Some models are used for reasons of size or ease rather than the true modeling of a physiological process. Application to granting agencies and design of animal studies is difficult without standardization of the ideal preclinical model for disease states. The current article addresses the preclinical animal modeling of osteoporosis, infection, bone defects, and cartilage injury. This article is a discussion of the current literature, commonly used models, and suggests preferred preclinical models for future research design.


Assuntos
Pesquisa Biomédica/tendências , Modelos Animais de Doenças , Traumatologia/tendências , Ferimentos e Lesões/diagnóstico , Ferimentos e Lesões/terapia , Animais , Humanos , Ferimentos e Lesões/veterinária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA