Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Knee Surg Sports Traumatol Arthrosc ; 28(10): 3245-3257, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31894366

RESUMO

PURPOSE: To systematically analyse post-operative outcomes following enhanced microfracture procedures in focal cartilage injuries of the knee. METHODS: Database searches were conducted in PubMed, EMBASE and Cochrane Library databases up to 30 November 2018, for clinical studies in humans that assessed surgical outcomes of enhanced microfracture procedures in focal cartilage injuries of the knee. The clinical, functional and imaging outcomes were assessed and summarized. The MINORS scale was used to assess the methodological quality of the studies included. RESULTS: Ten studies were included comprising a total of 331 patients (mean age of 37.0 ± 5.5 years, body mass 25.2 ± 1.7 kg m2, 56% male and 42% left knee), 278 femoral condyle chondral defects (147 medial, 35 lateral and 78 undefined) and 43 chondral defects distributed by the tibial plateau, patella and femoral trochlea. The chondral defects were mostly Outerbridge grade III or IV and the mean defect size was 3.2 ± 0.6 cm2. Studies consistently demonstrated significant improvement in the patient-reported outcome measures from baseline to final follow-up. Overall, imaging outcomes showed inconsistent results. Treatment-related adverse events were poorly reported. CONCLUSION: Enhanced microfracture techniques significantly result in improved patient-reported outcome measures over the MCID, but provide inconsistent imaging results. Current clinical evidence does not allow for unequivocal recommendation of enhanced microfracture to treat symptomatic focal grade III/IV knee cartilage lesions. LEVEL OF EVIDENCE: IV.


Assuntos
Artroplastia Subcondral/métodos , Cartilagem Articular/lesões , Cartilagem Articular/cirurgia , Traumatismos do Joelho/cirurgia , Alicerces Teciduais , Adulto , Feminino , Humanos , Masculino , Medidas de Resultados Relatados pelo Paciente , Resultado do Tratamento
2.
Pediatr Infect Dis J ; 38(5): 453-458, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30346369

RESUMO

BACKGROUND: Infective endocarditis (IE) remains a diagnostic and therapeutic challenge associated with high morbidity and mortality. We evaluated the microbial profile and clinical manifestation of IE in children. METHODS: A retrospective study examining pediatric IE cases treated between 2000 and 2017 at the Department of Pediatric Cardiology, KU Leuven, was conducted. Clinical presentation, treatment, complications, outcome of IE, underlying microorganisms and congenital heart defects were reviewed. RESULTS: Fifty-three patients were diagnosed with IE. Overall, 19 patients (36%) required cardiac surgery. Seven patients (13%) died. Eighty-seven percent of patients had an underlying congenital cardiac defect. Eighteen (34%) children presented with prosthetic graft IE. A causative organism was found in 49 (92%) cases: viridans group streptococci were identified in 17 (32%), Staphylococcus aureus in 13 (25%) and coagulase-negative staphylococci in 11 (20%) children. Community-acquired (CA) IE increased significantly from 8 (33%) cases in 2000-2007 to 20 (74%) cases in 2008-2017 (P < 0.01). Even with viridans streptococci being significantly more prevalent in the CA group (P < 0.01), we did not observe an increase of streptococcal IE from 2008 to 2017. Seventeen (32%) patients presented with hospital-acquired IE during the first year of life with 14 (82%) children after surgery and a prevalence of coagulase-negative staphylococci (53%). CONCLUSIONS: The incidence of pediatric IE was similar over the investigated time period with a shift toward CA IE. Streptococci and staphylococci accounted for the majority of cases in both periods. Awareness of IE and its prevention is crucial in patients after implantation of prosthetic grafts.


Assuntos
Bactérias/isolamento & purificação , Infecções Bacterianas/microbiologia , Infecções Bacterianas/patologia , Endocardite/microbiologia , Endocardite/patologia , Adolescente , Bactérias/classificação , Infecções Bacterianas/mortalidade , Infecções Bacterianas/terapia , Bélgica/epidemiologia , Criança , Pré-Escolar , Endocardite/mortalidade , Endocardite/terapia , Feminino , Hospitais Pediátricos , Hospitais Universitários , Humanos , Lactente , Recém-Nascido , Masculino , Prevalência , Estudos Retrospectivos , Análise de Sobrevida , Resultado do Tratamento
3.
Virulence ; 9(1): 1615-1624, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30280967

RESUMO

Staphylococcus aureus is the leading cause of infective endocarditis (IE). While the role of S. aureus cell-wall associated protein clumping factor A (ClfA) in promoting IE has been already demonstrated, that of the secreted plasma-clotting factors staphylocoagulase (Coa) and von Willebrand factor-binding protein (vWbp) has not yet been elucidated. We investigated the role of Coa and vWbp in IE initiation in rats with catheter-induced aortic vegetations, using Lactococcus lactis expressing coa, vWbp, clfA or vWbp/clfA, and S. aureus Newman Δcoa, ΔvWbp, ΔclfA or Δcoa/ΔvWbp/ΔclfA mutants. vWbp-expression increased L. lactis valve infection compared to parent and coa-expressing strains (incidence: 62%, versus 0% and 13%, respectively; P < 0.01). Likewise, expression of clfA increased L. lactis infectivity (incidence: 80%), which was not further affected by co-expression of vWbp. In symmetry, deletion of the coa or vWbp genes in S. aureus did not decrease infectivity (incidence: 68 and 64%, respectively) whereas deletion of clfA did decrease valve infection (incidence: 45%; P = 0.03 versus parent), which was not further affected by the triple deletion Δcoa/ΔvWbp/ΔclfA (incidence: 36%; P > 0.05 versus ΔclfA mutant). Coa does not support the initial colonization of IE (in L. lactis) without other key virulence factors and vWbp contributes to initiation of IE (in L. lactis) but is marginal in the present of ClfA.


Assuntos
Valva Aórtica/microbiologia , Proteínas de Bactérias/metabolismo , Coagulase/metabolismo , Endocardite Bacteriana/patologia , Staphylococcus aureus/genética , Fator de von Willebrand/metabolismo , Animais , Valva Aórtica/fisiopatologia , Proteínas de Bactérias/genética , Infecções Relacionadas a Cateter/microbiologia , Coagulase/genética , Feminino , Deleção de Genes , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Ratos , Ratos Wistar , Infecções Estafilocócicas , Staphylococcus aureus/patogenicidade , Fatores de Virulência/genética
5.
Thromb Haemost ; 118(7): 1230-1241, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29909601

RESUMO

Adhesion of Staphylococcus aureus to endothelial cells (ECs) is paramount in infective endocarditis. Bacterial proteins such as clumping factor A (ClfA) and fibronectin binding protein A (FnbpA) mediate adhesion to EC surface molecules and (sub)endothelial matrix proteins including fibrinogen (Fg), fibrin, fibronectin (Fn) and von Willebrand factor (vWF). We studied the influence of shear flow and plasma on the binding of ClfA and FnbpA (including its sub-domains A, A16+, ABC, CD) to coverslip-coated vWF, Fg/fibrin, Fn or confluent ECs, making use of Lactococcus lactis, expressing these adhesins heterologously. Global adherence profiles were similar in static and flow conditions. In the absence of plasma, L. lactis-clfA binding to Fg increased with shear forces, whereas binding to fibrin did not. The degree of adhesion of L. lactis-fnbpA to EC-bound Fn and of L. lactis-clfA to EC-bound Fg, furthermore, was similar to that of L. lactis-clfA to coated vWF domain A1, in the presence of vWF-binding protein (vWbp). Yet, in plasma, L. lactis-clfA adherence to activated EC-vWF/vWbp dropped over 10 minutes by 80% due to vWF-hydrolysis by a disintegrin and metalloproteinase with thrombospondin type 1 motif, member 13 and that of L. lactis-fnbpA likewise by > 70% compared to the adhesion in absence of plasma. In contrast, plasma Fg supported high L. lactis-clfA binding to resting and activated ECs. Or, in plasma S. aureus adhesion to active endothelium occurs mainly via two complementary pathways: a rapid but short-lived vWF/vWbp pathway and a stable integrin-coupled Fg-pathway. Hence, the pharmacological inhibition of ClfA-Fg interactions may constitute a valuable additive treatment in infective endocarditis.


Assuntos
Proteína ADAMTS13/sangue , Aderência Bacteriana , Coagulase/metabolismo , Endocardite Bacteriana/microbiologia , Células Endoteliais da Veia Umbilical Humana/microbiologia , Plasma/enzimologia , Staphylococcus aureus/metabolismo , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Células Cultivadas , Coagulase/genética , Endocardite Bacteriana/sangue , Fibrina/metabolismo , Fibrinogênio , Fibronectinas/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Staphylococcus aureus/genética , Estresse Mecânico , Fator de von Willebrand/metabolismo
6.
J Thorac Cardiovasc Surg ; 155(1): 325-332.e4, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28712577

RESUMO

BACKGROUND: Various conduits and stent-mounted valves are used as pulmonary valve graft tissues for right ventricular outflow tract reconstruction with good hemodynamic results. Valve replacement carries an increased risk of infective endocarditis (IE). Recent observations have increased awareness of the risk of IE after transcatheter implantation of a stent-mounted bovine jugular vein valve. This study focused on the susceptibility of graft tissue surfaces to bacterial adherence as a potential risk factor for subsequent IE. METHODS: Adhesion of Staphylococcus aureus, Staphylococcus epidermidis, and Streptococcus sanguinis to bovine pericardium (BP) patch, bovine jugular vein (BJV), and cryopreserved homograft (CH) tissues was quantified under static and shear stress conditions. Microscopic analysis and histology were performed to evaluate bacterial adhesion to matrix components. RESULTS: In general, similar bacteria numbers were recovered from CH and BJV tissue surfaces for all strains, especially in flow conditions. Static bacterial adhesion to the CH wall was lower for S sanguinis adhesion (P < .05 vs BP patch). Adhesion to the BJV wall, CH wall, and leaflet was decreased for S epidermidis in static conditions (P < .05 vs BP patch). Bacterial adhesion under shear stress indicated similar bacterial adhesion to all tissues, except for lower adhesion to the BJV wall after S sanguinis incubation. Microscopic analysis showed the importance of matrix component exposure for bacterial adherence to CH. CONCLUSIONS: Our data provide evidence that the surface composition of BJV and CH tissues themselves, bacterial surface proteins, and shear forces per se are not the prime determinants of bacterial adherence.


Assuntos
Aderência Bacteriana/fisiologia , Bioprótese , Endocardite Bacteriana , Implante de Prótese de Valva Cardíaca/efeitos adversos , Próteses Valvulares Cardíacas , Infecções Estafilocócicas , Staphylococcus , Animais , Bioprótese/efeitos adversos , Bioprótese/microbiologia , Bovinos , Endocardite Bacteriana/etiologia , Endocardite Bacteriana/prevenção & controle , Próteses Valvulares Cardíacas/efeitos adversos , Próteses Valvulares Cardíacas/microbiologia , Implante de Prótese de Valva Cardíaca/métodos , Humanos , Veias Jugulares/transplante , Infecções Relacionadas à Prótese/etiologia , Infecções Relacionadas à Prótese/prevenção & controle , Valva Pulmonar/cirurgia , Infecções Estafilocócicas/etiologia , Infecções Estafilocócicas/prevenção & controle , Staphylococcus/classificação , Staphylococcus/fisiologia , Propriedades de Superfície , Válvulas Venosas/transplante , Obstrução do Fluxo Ventricular Externo/cirurgia
7.
J Infect Dis ; 213(7): 1148-56, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26743845

RESUMO

BACKGROUND: Staphylococcus lugdunensis is an emerging cause of endocarditis. To cause endovascular infections, S. lugdunensis requires mechanisms to overcome shear stress. We investigated whether platelets and von Willebrand factor (VWF) mediate bacterial adhesion to the vessel wall and the cardiac valves under flow. METHODS: S. lugdunensis binding to VWF, collagen, and endothelial cells was studied in a parallel flow chamber in the absence and presence of platelets. In vivo adhesion of S. lugdunensis was evaluated in a mouse microvasculature perfusion model and a new mouse model of endocarditis. RESULTS: Contrary to other coagulase-negative staphylococci, S. lugdunensis bound to VWF under flow, thus enabling its adhesion to endothelial cells and to the subendothelial matrix. In inflamed vessels of the mesenteric circulation, VWF recruited S. lugdunensis to the vessel wall. In a novel endocarditis mouse model, local inflammation and the resulting release of VWF enabled S. lugdunensis to bind and colonize the heart valves. CONCLUSIONS: S. lugdunensis binds directly to VWF, which proved to be vital for withstanding shear forces and for its adhesion to the vessel wall and cardiac valves. This mechanism explains why S. lugdunensis causes more-aggressive infections, including endocarditis, compared with other coagulase-negative staphylococci.


Assuntos
Aderência Bacteriana/fisiologia , Endocardite Bacteriana/microbiologia , Valvas Cardíacas/microbiologia , Infecções Estafilocócicas/microbiologia , Staphylococcus lugdunensis/fisiologia , Fator de von Willebrand/metabolismo , Animais , Regulação da Expressão Gênica , Humanos , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica , Resistência ao Cisalhamento , Fator de von Willebrand/genética
8.
Pathog Dis ; 73(8): ftv060, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26310867

RESUMO

Enterococcus faecalis and Streptococcus gallolyticus cause infective endocarditis (IE), which can originate from the continuous release or translocation of low bacterial numbers into the bloodstream. In this context, IE cannot be prevented with antibiotics. We previously demonstrated that aspirin plus ticlopidine protected rats from IE due to S. gordonii and Staphylococcus aureus. Here we showed that aspirin plus ticlopidine significantly reduced vegetation weight and protected 73 and 64% rats (P < 0.005) from IE due to E. faecalis and S. gallolyticus, respectively. These results further support the potential use of aspirin plus ticlopidine for a global prevention of IE in high-risk patients.


Assuntos
Aspirina/administração & dosagem , Endocardite Bacteriana/prevenção & controle , Enterococcus faecalis/crescimento & desenvolvimento , Infecções por Bactérias Gram-Positivas/prevenção & controle , Inibidores da Agregação Plaquetária/farmacologia , Streptococcus/crescimento & desenvolvimento , Ticlopidina/administração & dosagem , Animais , Modelos Animais de Doenças , Endocardite Bacteriana/microbiologia , Feminino , Infecções por Bactérias Gram-Positivas/microbiologia , Ratos Wistar , Resultado do Tratamento
9.
Vaccine ; 33(30): 3512-7, 2015 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-26048778

RESUMO

Staphylococcus aureus is a major cause of serious infections in humans and animals and a vaccine is becoming a necessity. Lactococcus lactis is a non-pathogenic bacterium that can be used as a vector for the delivery of antigens. We investigated the ability of non-living L. lactis heterologously expressing S. aureus clumping factor A (ClfA) and fibronectin-binding protein A (FnbpA), alone or together, to elicit an immune response in rats and protect them from S. aureus experimental infective endocarditis (IE). L. lactis ClfA was used for immunization against S. aureus Newman (expressing ClfA but not FnbpA), while L. lactis ClfA, L. lactis FnbpA, as well as L. lactis ClfA/FnbpA, were used against S. aureus P8 (expressing ClfA and FnbpA). Vaccination of rats with L. lactis ClfA elicited antibodies that inhibited binding of S. aureus Newman to fibrinogen, triggered the production of IL-17A and conferred protection to 13/19 (68%) of the animals from IE (P<0.05). Immunization with L. lactis ClfA, L. lactis FnbpA or L. lactis ClfA/FnbpA also produced antibodies against the target proteins, but these did not prevent binding of S. aureus P8 to fibrinogen or fibronectin and did not protect animals against S. aureus P8 IE. Moreover, immunization with constructs containing FnbpA did not increase IL-17A production. These results indicate that L. lactis is a valuable antigen delivery system able to elicit efficient humoral and cellular responses. However, the most appropriate antigens affording protection against S. aureus IE are yet to be elucidated.


Assuntos
Adesinas Bacterianas/imunologia , Coagulase/imunologia , Portadores de Fármacos , Endocardite/prevenção & controle , Lactococcus lactis/genética , Vacinas Antiestafilocócicas/imunologia , Staphylococcus aureus/imunologia , Adesinas Bacterianas/genética , Animais , Anticorpos Antibacterianos/sangue , Coagulase/genética , Modelos Animais de Doenças , Endocardite/imunologia , Feminino , Fibronectinas/metabolismo , Ratos Wistar , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Vacinas Antiestafilocócicas/administração & dosagem , Vacinas Antiestafilocócicas/genética , Staphylococcus aureus/genética , Resultado do Tratamento , Vacinação/métodos
11.
J Infect Dis ; 211(1): 72-9, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25086177

RESUMO

BACKGROUND: Infective endocarditis (IE) mostly occurs after spontaneous low-grade bacteremia. Thus, IE cannot be prevented by circumstantial antibiotic prophylaxis. Platelet activation following bacterial-fibrinogen interaction or thrombin-mediated fibrinogen-fibrin polymerization is a critical step in vegetation formation. We tested the efficacy of antiplatelet and antithrombin to prevent experimental IE. METHODS: A rat model of experimental IE following prolonged low-grade bacteremia mimicking smoldering bacteremia in humans was used. Prophylaxis with antiplatelets (aspirin, ticlopidine [alone or in combination], eptifibatide, or abciximab) or anticoagulants (antithrombin dabigatran etexilate or anti-vitamin K acenocoumarol) was started 2 days before inoculation with Streptococcus gordonii or Staphylococcus aureus. Valve infection was assessed 24 hours later. RESULTS: Aspirin plus ticlopidine, as well as abciximab, protected 45%-88% of animals against S. gordonii and S. aureus IE (P < .05). Dabigatran etexilate protected 75% of rats against IE due to S. aureus (P < .005) but failed to protect against S. gordonii (<30% protection). Acenocoumarol was ineffective. CONCLUSIONS: Antiplatelet and direct antithrombin agents may be useful in the prophylaxis of IE in humans. In particular, the potential dual benefit of dabigatran etexilate might be reconsidered for patients with prosthetic valves, who require life-long anticoagulation and in whom S. aureus IE is associated with high mortality.


Assuntos
Endocardite Bacteriana/tratamento farmacológico , Fibrinolíticos/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Animais , Antibioticoprofilaxia/métodos , Bacteriemia/tratamento farmacológico , Bacteriemia/microbiologia , Modelos Animais de Doenças , Endocardite Bacteriana/microbiologia , Humanos , Ratos , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/efeitos dos fármacos , Streptococcus gordonii/efeitos dos fármacos
12.
Int J Med Microbiol ; 303(8): 498-504, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23856340

RESUMO

Streptococcus tigurinus is responsible for systemic infections in humans including infective endocarditis. We investigated whether the invasive trait of S. tigurinus in humans correlated with an increased ability to induce IE in rats. Rats with catheter-induced aortic vegetations were inoculated with 104 CFU/ml of either of four S. tigurinus strains AZ_3a(T), AZ_4a, AZ_8 and AZ_14, isolated from patients with infective endocarditis or with the well known IE pathogen Streptococcus gordonii (Challis). Aortic infection was assessed after 24 h. S. tigurinus AZ_3a(T), AZ_4a and AZ_14 produced endocarditis in ≥80% of rats whereas S. gordonii produced endocarditis in only 33% of animals (P<0.05). S. tigurinus AZ_8 caused vegetation infection in 56% of the animals. The capacity of S. tigurinus to induce aortic infection was not related to their ability to bind extracellular matrix proteins (fibrinogen, fibronectin or collagen) or to trigger platelet aggregation. However, all S. tigurinus isolates showed an enhanced resistance to phagocytosis by macrophages and two of them had an increased ability to enter endothelial cells, key attributes of invasive streptococcal species.


Assuntos
Modelos Animais de Doenças , Endocardite Bacteriana/microbiologia , Endocardite Bacteriana/patologia , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/patologia , Streptococcus/patogenicidade , Animais , Aorta/microbiologia , Aorta/patologia , Células Endoteliais/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Fagocitose , Ratos , Streptococcus/imunologia
13.
Infect Immun ; 81(3): 697-703, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23250949

RESUMO

Animal models of infective endocarditis (IE) induced by high-grade bacteremia revealed the pathogenic roles of Staphylococcus aureus surface adhesins and platelet aggregation in the infection process. In humans, however, S. aureus IE possibly occurs through repeated bouts of low-grade bacteremia from a colonized site or intravenous device. Here we used a rat model of IE induced by continuous low-grade bacteremia to explore further the contributions of S. aureus virulence factors to the initiation of IE. Rats with aortic vegetations were inoculated by continuous intravenous infusion (0.0017 ml/min over 10 h) with 10(6) CFU of Lactococcus lactis pIL253 or a recombinant L. lactis strain expressing an individual S. aureus surface protein (ClfA, FnbpA, BCD, or SdrE) conferring a particular adhesive or platelet aggregation property. Vegetation infection was assessed 24 h later. Plasma was collected at 0, 2, and 6 h postinoculation to quantify the expression of tumor necrosis factor (TNF), interleukin 1α (IL-1α), IL-1ß, IL-6, and IL-10. The percentage of vegetation infection relative to that with strain pIL253 (11%) increased when binding to fibrinogen was conferred on L. lactis (ClfA strain) (52%; P = 0.007) and increased further with adhesion to fibronectin (FnbpA strain) (75%; P < 0.001). Expression of fibronectin binding alone was not sufficient to induce IE (BCD strain) (10% of infection). Platelet aggregation increased the risk of vegetation infection (SdrE strain) (30%). Conferring adhesion to fibrinogen and fibronectin favored IL-1ß and IL-6 production. Our results, with a model of IE induced by low-grade bacteremia, resembling human disease, extend the essential role of fibrinogen binding in the initiation of S. aureus IE. Triggering of platelet aggregation or an inflammatory response may contribute to or promote the development of IE.


Assuntos
Adesinas Bacterianas/metabolismo , Bacteriemia/imunologia , Endocardite Bacteriana/imunologia , Agregação Plaquetária/imunologia , Staphylococcus aureus/metabolismo , Adesinas Bacterianas/genética , Animais , Aderência Bacteriana/fisiologia , Citocinas/genética , Citocinas/metabolismo , Fibrinogênio , Fibronectinas , Regulação da Expressão Gênica/imunologia , Humanos , Proteínas Imobilizadas , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...