Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 43(2): 267-285, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36453281

RESUMEN

BACKGROUND: Infective endocarditis (IE) is characterized by an infected thrombus at the heart valves. How bacteria bypass the immune system and cause these thrombi remains unclear. Neutrophils releasing NETs (neutrophil extracellular traps) lie at this interface between host defense and coagulation. We aimed to determine the role of NETs in IE immunothrombosis. METHODS: We used a murine model of Staphylococcus aureus endocarditis in which IE is provoked on inflamed heart valves and characterized IE thrombus content by immunostaining identifying NETs. Antibody-mediated neutrophil depletion and neutrophil-selective PAD4 (peptidylarginine deiminase 4)-knockout mice were used to clarify the role of neutrophils and NETs, respectively. S. aureus mutants deficient in key virulence factors related to immunothrombosis (nucleases or staphylocoagulases) were investigated. RESULTS: Neutrophils releasing NETs were present in infected thrombi and within cellular infiltrates in the surrounding vasculature. Neutrophil depletion increased occurrence of IE, whereas neutrophil-selective impairment of NET formation did not alter IE occurrence. Absence of S. aureus nuclease, which degrades NETs, did not affect endocarditis outcome. In contrast, absence of staphylocoagulases (coagulase and von Willebrand factor binding protein) led to improved survival, decreased bacteremia, smaller infiltrates, and decreased tissue destruction. Significantly more NETs were present in these vegetations, which correlated with decreased bacteria and cell death in the adjacent vascular wall. CONCLUSIONS: Neutrophils protect against IE independent of NET release. Absence of S. aureus coagulases, but not nucleases, reduced IE severity and increased NET levels. Staphylocoagulase-induced fibrin likely hampers NETs from constraining infection and the resultant tissue damage, a hallmark of valve destruction in IE.


Asunto(s)
Endocarditis Bacteriana , Endocarditis , Trampas Extracelulares , Infecciones Estafilocócicas , Ratones , Animales , Neutrófilos/metabolismo , Trampas Extracelulares/metabolismo , Staphylococcus aureus , Tromboinflamación , Endocarditis Bacteriana/prevención & control , Endocarditis Bacteriana/metabolismo , Endocarditis/metabolismo
2.
Glycobiology ; 31(11): 1582-1595, 2021 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-34459483

RESUMEN

Streptococcus gordonii and Streptococcus sanguinis are primary colonizers of tooth surfaces and are generally associated with oral health, but can also cause infective endocarditis (IE). These species express "Siglec-like" adhesins that bind sialylated glycans on host glycoproteins, which can aid the formation of infected platelet-fibrin thrombi (vegetations) on cardiac valve surfaces. We previously determined that the ability of S. gordonii to bind sialyl T-antigen (sTa) increased pathogenicity, relative to recognition of sialylated core 2 O-glycan structures, in an animal model of IE. However, it is unclear when and where the sTa structure is displayed, and which sTa-modified host factors promote valve colonization. In this study, we identified sialylated glycoproteins in the aortic valve vegetations and plasma of rat and rabbit models of this disease. Glycoproteins that display sTa vs. core 2 O-glycan structures were identified by using recombinant forms of the streptococcal Siglec-like adhesins for lectin blotting and affinity capture, and the O-linked glycans were profiled by mass spectrometry. Proteoglycan 4 (PRG4), also known as lubricin, was a major carrier of sTa in the infected vegetations. Moreover, plasma PRG4 levels were significantly higher in animals with damaged or infected valves, as compared with healthy animals. The combined results demonstrate that, in addition to platelet GPIbα, PRG4 is a highly sialylated mucin-like glycoprotein found in aortic valve vegetations and may contribute to the persistence of oral streptococci in this protected endovascular niche. Moreover, plasma PRG4 could serve as a biomarker for endocardial injury and infection.


Asunto(s)
Modelos Animales de Enfermedad , Endocarditis Bacteriana/metabolismo , Válvulas Cardíacas/metabolismo , Proteoglicanos/metabolismo , Streptococcus gordonii/aislamiento & purificación , Animales , Endocarditis Bacteriana/microbiología , Endocarditis Bacteriana/patología , Femenino , Válvulas Cardíacas/microbiología , Válvulas Cardíacas/patología , Humanos , Conejos , Ratas , Ratas Sprague-Dawley
3.
PLoS Pathog ; 17(1): e1009222, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33465168

RESUMEN

Bacterial binding to platelets is a key step in the development of infective endocarditis (IE). Sialic acid, a common terminal carbohydrate on host glycans, is the major receptor for streptococci on platelets. So far, all defined interactions between streptococci and sialic acid on platelets are mediated by serine-rich repeat proteins (SRRPs). However, we identified Streptococcus oralis subsp. oralis IE-isolates that bind sialic acid but lack SRRPs. In addition to binding sialic acid, some SRRP- isolates also bind the cryptic receptor ß-1,4-linked galactose through a yet unknown mechanism. Using comparative genomics, we identified a novel sialic acid-binding adhesin, here named AsaA (associated with sialic acid adhesion A), present in IE-isolates lacking SRRPs. We demonstrated that S. oralis subsp. oralis AsaA is required for binding to platelets in a sialic acid-dependent manner. AsaA comprises a non-repeat region (NRR), consisting of a FIVAR/CBM and two Siglec-like and Unique domains, followed by 31 DUF1542 domains. When recombinantly expressed, Siglec-like and Unique domains competitively inhibited binding of S. oralis subsp. oralis and directly interacted with sialic acid on platelets. We further demonstrated that AsaA impacts the pathogenesis of S. oralis subsp. oralis in a rabbit model of IE. Additionally, we found AsaA orthologues in other IE-causing species and demonstrated that the NRR of AsaA from Gemella haemolysans blocked binding of S. oralis subsp. oralis, suggesting that AsaA contributes to the pathogenesis of multiple IE-causing species. Finally, our findings provide evidence that sialic acid is a key factor for bacterial-platelets interactions in a broader range of species than previously appreciated, highlighting its potential as a therapeutic target.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Endocarditis Bacteriana/patología , Ácido N-Acetilneuramínico/metabolismo , Streptococcus/metabolismo , Adhesinas Bacterianas/genética , Animales , Proteínas Bacterianas/genética , Endocarditis Bacteriana/metabolismo , Endocarditis Bacteriana/microbiología , Masculino , Conejos , Streptococcus/clasificación , Streptococcus/genética , Streptococcus/aislamiento & purificación
4.
J Nucl Cardiol ; 28(6): 2533-2542, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-32043240

RESUMEN

PURPOSE: This study aimed at determining the diagnostic implications of indirect signs of infection at FDG-PET-i.e., hypermetabolisms of the spleen and/or bone marrow (HSBM)-when documented in patients with known or suspected infective endocarditis (IE). METHODS: HSBM were defined by higher mean standardized uptake values comparatively to that of the liver on FDG-PET images from patients with a high likelihood of IE and prospectively included in a multicenter study. RESULTS: Among the 129 included patients, IE was ultimately deemed as definite in 88 cases. HSBM was a predictor of definite IE (P = 0.014; odds ratio (OR) 3.2), independently of the criterion of an abnormal cardiac FDG uptake (P = 0.0007; OR 9.68), and a definite IE was documented in 97% (29/30) of patients showing both HSBM and abnormal cardiac uptake, 78% (7/9) of patients with only abnormal cardiac uptake, 67% (42/63) of patients with only HSBM, and 37% (10/27) of patients with neither one. CONCLUSION: In this cohort with a high likelihood of IE, HSBM is an additional albeit indirect sign of IE, independently of the criterion of an abnormal cardiac uptake, and could reinforce the suspicion of IE in the absence of any other infectious, inflammatory, or malignant disease.


Asunto(s)
Médula Ósea/metabolismo , Endocarditis Bacteriana/diagnóstico por imagen , Endocarditis Bacteriana/metabolismo , Fluorodesoxiglucosa F18 , Tomografía de Emisión de Positrones , Radiofármacos , Bazo/metabolismo , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Tomografía de Emisión de Positrones/métodos , Estudios Prospectivos
5.
PLoS One ; 15(12): e0243365, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33290425

RESUMEN

The combination of ampicillin (AMP) and ceftriaxone (CRO) is considered synergistic against Enterococcus faecalis based on in vitro tests and the rabbit endocarditis model, however, in vitro assays are limited by the use of fixed antibiotic concentrations and the rabbit model by poor bacterial growth, high variability, and the use of point dose-effect estimations, that may lead to inaccurate assessment of antibiotic combinations and hinder optimal translation. Here, we tested AMP+CRO against two strains of E. faecalis and one of E. faecium in an optimized mouse thigh infection model that yields high bacterial growth and allows to define the complete dose-response relationship. By fitting Hill's sigmoid model and estimating the parameters maximal effect (Emax) and effective dose 50 (ED50), the following interactions were defined: synergism (Emax increase ≥2 log10 CFU/g), antagonism (Emax reduction ≥1 log10 CFU/g) and potentiation (ED50 reduction ≥50% without changes in Emax). AMP monotherapy was effective against the three strains, yielding valid dose-response curves in terms of dose and the index fT>MIC. CRO monotherapy showed no effect. The combination AMP+CRO against E. faecalis led to potentiation (59-81% ED50 reduction) and not synergism (no changes in Emax). Against E. faecium, the combination was indifferent. The optimized mouse infection model allowed to obtain the complete dose-response curve of AMP+CRO and to define its interaction based on pharmacodynamic parameter changes. Integrating these results with the pharmacokinetics will allow to derive the PK/PD index bound to the activity of the combination, essential for proper translation to the clinic.


Asunto(s)
Ampicilina , Ceftriaxona , Endocarditis Bacteriana , Enterococcus faecalis/metabolismo , Enterococcus faecium/metabolismo , Infecciones por Bacterias Grampositivas , Ampicilina/farmacocinética , Ampicilina/farmacología , Animales , Ceftriaxona/farmacocinética , Ceftriaxona/farmacología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Endocarditis Bacteriana/tratamiento farmacológico , Endocarditis Bacteriana/metabolismo , Endocarditis Bacteriana/microbiología , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Infecciones por Bacterias Grampositivas/metabolismo , Infecciones por Bacterias Grampositivas/microbiología , Ratones , Conejos
7.
Eur Heart J ; 40(39): 3248-3259, 2019 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-30945735

RESUMEN

AIMS: The pathogenesis of endocarditis is not well understood resulting in unsuccessful attempts at prevention. Clinical observations suggest that Staphylococcus aureus infects either damaged or inflamed heart valves. Using a newly developed endocarditis mouse model, we therefore studied the initial adhesion of S. aureus in both risk states. METHODS AND RESULTS: Using 3D confocal microscopy, we examined the adhesion of fluorescent S. aureus to murine aortic valves. To mimic different risk states we either damaged the valves with a surgically placed catheter or simulated valve inflammation by local endothelium activation. We used von Willebrand factor (VWF) gene-deficient mice, induced platelet and fibrinogen depletion and used several S. aureus mutant strains to investigate the contribution of both host and bacterial factors in early bacterial adhesion. Both cardiac valve damage and inflammation predisposed to endocarditis, but by distinct mechanisms. Following valve damage, S. aureus adhered directly to VWF and fibrin, deposited on the damaged valve. This was mediated by Sortase A-dependent adhesins such as VWF-binding protein and Clumping factor A. Platelets did not contribute. In contrast, upon cardiac valve inflammation, widespread endothelial activation led to endothelial cell-bound VWF release. This recruited large amounts of platelets, capturing S. aureus to the valve surface. Here, neither fibrinogen, nor Sortase A were essential. CONCLUSION: Cardiac valve damage and inflammation predispose to S. aureus endocarditis via distinct mechanisms. These findings may have important implications for the development of new preventive strategies, as some interventions might be effective in one risk state, but not in the other.


Asunto(s)
Válvula Aórtica/microbiología , Adhesión Bacteriana , Endocarditis Bacteriana/microbiología , Inflamación/complicaciones , Infecciones Estafilocócicas/complicaciones , Staphylococcus aureus/fisiología , Animales , Válvula Aórtica/lesiones , Plaquetas , Coagulasa/metabolismo , Modelos Animales de Enfermedad , Endocarditis Bacteriana/metabolismo , Endotelio/metabolismo , Femenino , Fibrina/metabolismo , Inflamación/metabolismo , Masculino , Ratones , Glicoproteínas de Membrana Plaquetaria/metabolismo , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/metabolismo , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo
8.
PLoS Pathog ; 14(9): e1007102, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30235334

RESUMEN

Manganese (Mn) is an essential micronutrient that is not readily available to pathogens during infection due to an active host defense mechanism known as nutritional immunity. To overcome this nutrient restriction, bacteria utilize high-affinity transporters that allow them to compete with host metal-binding proteins. Despite the established role of Mn in bacterial pathogenesis, little is known about the relevance of Mn in the pathophysiology of E. faecalis. Here, we identified and characterized the major Mn acquisition systems of E. faecalis. We discovered that the ABC-type permease EfaCBA and two Nramp-type transporters, named MntH1 and MntH2, work collectively to promote cell growth under Mn-restricted conditions. The simultaneous inactivation of EfaCBA, MntH1 and MntH2 (ΔefaΔmntH1ΔmntH2 strain) led to drastic reductions (>95%) in cellular Mn content, severe growth defects in body fluids (serum and urine) ex vivo, significant loss of virulence in Galleria mellonella, and virtually complete loss of virulence in rabbit endocarditis and murine catheter-associated urinary tract infection (CAUTI) models. Despite the functional redundancy of EfaCBA, MntH1 and MntH2 under in vitro or ex vivo conditions and in the invertebrate model, dual inactivation of efaCBA and mntH2 (ΔefaΔmntH2 strain) was sufficient to prompt maximal sensitivity to calprotectin, a Mn- and Zn-chelating host antimicrobial protein, and for the loss of virulence in mammalian models. Interestingly, EfaCBA appears to play a prominent role during systemic infection, whereas MntH2 was more important during CAUTI. The different roles of EfaCBA and MntH2 in these sites could be attributed, at least in part, to the differential expression of efaA and mntH2 in cells isolated from hearts or from bladders. Collectively, this study demonstrates that Mn acquisition is essential for the pathogenesis of E. faecalis and validates Mn uptake systems as promising targets for the development of new antimicrobials.


Asunto(s)
Enterococcus faecalis/metabolismo , Enterococcus faecalis/patogenicidad , Manganeso/metabolismo , Virulencia/fisiología , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Infecciones Relacionadas con Catéteres/etiología , Infecciones Relacionadas con Catéteres/metabolismo , Infecciones Relacionadas con Catéteres/microbiología , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Modelos Animales de Enfermedad , Endocarditis Bacteriana/etiología , Endocarditis Bacteriana/metabolismo , Endocarditis Bacteriana/microbiología , Enterococcus faecalis/genética , Infecciones por Bacterias Grampositivas/etiología , Infecciones por Bacterias Grampositivas/metabolismo , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Complejo de Antígeno L1 de Leucocito/metabolismo , Ratones , Mariposas Nocturnas/metabolismo , Mariposas Nocturnas/microbiología , Conejos , Infecciones Urinarias/etiología , Infecciones Urinarias/metabolismo , Infecciones Urinarias/microbiología
9.
Microbiology (Reading) ; 164(10): 1254-1265, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30091695

RESUMEN

The alarmone (p)ppGpp mediates the stringent response and has a recognized role in bacterial virulence. We previously reported a stringent response-like state in Enterococcus faecalis isolated from a rabbit foreign body abscess model and showed that E. faecalis mutants with varying levels of cellular (p)ppGpp [Δrel, ΔrelQ and the (p)ppGpp0 ΔrelΔrelQ] had differential abilities to persist within abscesses. In this study, we investigated whether (p)ppGpp contributes to the pathogenesis of E. faecalis infective endocarditis (IE), a biofilm infection of the heart valves. While the stringent response was not activated in heart valve-associated E. faecalis, deletion of the gene encoding the bifunctional (p)ppGpp synthetase/hydrolase Rel significantly impaired valve colonization. These results indicate that the presence of (p)ppGpp is dispensable for E. faecalis to cause IE, whereas the ability to regulate (p)ppGpp levels is critical for valve colonization. Next, we characterized how basal (p)ppGpp levels affect processes associated with IE pathogenesis. Despite being defective in binding to BSA-coated polystyrene surfaces, the Δrel strain bound to collagen- and fibronectin-coated surfaces and ex vivo porcine heart valves as well as the parent and ΔrelΔrelQ strains, ruling out the possibility that the impaired IE phenotype was due to an attachment defect. Moreover, differences in cellular (p)ppGpp levels did not affect extracellular gelatinase activity but significantly impaired enterococcal invasion of human coronary artery endothelial cells. Taken together, this study uncovers for the first time the fact that differences in basal (p)ppGpp levels, rather than the stringent response, differentially affect processes that contribute to the pathogenesis of IE.


Asunto(s)
Endocarditis Bacteriana/microbiología , Enterococcus faecalis/patogenicidad , Guanosina Pentafosfato/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Células Cultivadas , Modelos Animales de Enfermedad , Endocarditis Bacteriana/metabolismo , Endocarditis Bacteriana/patología , Células Endoteliales/metabolismo , Células Endoteliales/microbiología , Enterococcus faecalis/genética , Enterococcus faecalis/crecimiento & desarrollo , Enterococcus faecalis/metabolismo , Gelatinasas/metabolismo , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica , Humanos , Ligasas/genética , Ligasas/metabolismo , Conejos , Porcinos , Virulencia/genética
10.
Artículo en Inglés | MEDLINE | ID: mdl-30150459

RESUMEN

We tested the ability of clavulanic acid to restore the efficacy of cefazolin against Staphylococcus aureus TX0117, which exhibits the cefazolin inoculum effect (CzIE). In the rat infective endocarditis model, the coadministration of cefazolin plus clavulanic acid resulted in a significant reduction of bacterial counts (7.1 ± 0.5 log10 CFU/g) compared to that with cefazolin alone (2 ± 0.6 log10 CFU/g; P < 0.0001). The addition of a ß-lactamase inhibitor may be a viable strategy for overcoming the CzIE.


Asunto(s)
Cefazolina/farmacología , Ácido Clavulánico/farmacología , Endocarditis Bacteriana/tratamiento farmacológico , Endocarditis/tratamiento farmacológico , Meticilina/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Animales , Antibacterianos/farmacología , Endocarditis/metabolismo , Endocarditis Bacteriana/metabolismo , Pruebas de Sensibilidad Microbiana/métodos , Ratas , Infecciones Estafilocócicas/metabolismo , beta-Lactamasas/metabolismo
11.
Methods Mol Biol ; 1535: 229-241, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27914083

RESUMEN

In this protocol, we describe the application of using a high affinity monoclonal antibody generated against the major pilin protein component of the pilin structure of Enterococcus faecalis as a PET imaging agent for enterococcal endocarditis detection. The anti-pilin -mAb 64Cu conjugate was able to specifically label enterococcal endocarditis vegetation in vivo in a rodent endocarditis model. By targeting pili, a covalently linked surface antigen extending from the bacterial surface, we provided evidence that gram-positive pilin represent a logical surface antigen to define or target an infectious agent for molecularly guided imaging. Our goal in providing a detailed protocol of our efforts is to enable others to build upon this methodology to answer pertinent translational and basic research questions in the pursuit of diagnosis and treatment of infective endocarditis.


Asunto(s)
Anticuerpos Monoclonales , Endocarditis Bacteriana/diagnóstico por imagen , Endocarditis Bacteriana/metabolismo , Inmunoconjugados , Imagen Molecular/métodos , Animales , Biomarcadores , Modelos Animales de Enfermedad , Endocarditis Bacteriana/microbiología , Citometría de Flujo/métodos , Microscopía Electrónica , Tomografía de Emisión de Positrones , Ratas , Microtomografía por Rayos X
12.
Int J Cardiol ; 202: 564-70, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26447663

RESUMEN

Timely diagnosis of bacterial infective endocarditis (IE) is crucial, as mortality remains high in this severe bacterial infection, currently without any distinct biological markers. Our goal was to evaluate potential diagnostic biomarkers by reviewing current literature. The MEDLINE, Embase and Scopus databases were searched for articles published from 1980 through June 2015 restricted to English, Norwegian, Danish and Swedish. Eighteen studies qualified, providing a review of the most promising candidates for future studies. Several studies are inconclusive, since they are characterized by using improper control groups. Patients with IE have bacteremia, and control groups should therefore be patients with bacteremia without IE. Based on current research, N-terminal-pro-B-type natriuretic peptide (NT-proBNP) alone or in combination with Cystatin C (Cys C), lipopolysaccharide-binding protein (LBP), troponins, aquaporin-9 (AQP9), S100 calcium binding protein A11 (S100A11), E-selectin (CD62E) and VCAM-1 (CD54) and interleukin-6 (IL-6) are potential biomarkers for future studies.


Asunto(s)
Biomarcadores/metabolismo , Endocarditis Bacteriana/diagnóstico , Endocarditis/diagnóstico , Endocarditis/metabolismo , Endocarditis Bacteriana/metabolismo , Humanos
13.
Infect Immun ; 83(12): 4653-61, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26371130

RESUMEN

The interaction between bacteria and fibronectin is believed to play an important role in the pathogenicity of clinically important Gram-positive cocci. In the present study, we identified a gene encoding a predicted fibronectin-binding protein of Enterococcus faecium (fnm), a homologue of Streptococcus pneumoniae pavA, in the genomes of E. faecium strain TX82 and all other sequenced E. faecium isolates. Full-length recombinant Fnm from strain TX82 bound to immobilized fibronectin in a concentration-dependent manner and also appeared to bind collagen type V and laminin, but not other proteins, such as transferrin, heparin, bovine serum albumin, mucin, or collagen IV. We demonstrated that the N-terminal fragment of Fnm is required for full fibronectin binding, since truncation of this region caused a 2.4-fold decrease (P < 0.05) in the adhesion of E. faecium TX82 to fibronectin. Deletion of fnm resulted in a significant reduction (P < 0.001) in the ability of the mutant, TX6128, to bind fibronectin relative to that of the wild-type strain; in situ reconstitution of fnm in the deletion mutant strain restored adherence. In addition, the Δfnm mutant was highly attenuated relative to TX82 (P ≤ 0.0001) in a mixed-inoculum rat endocarditis model. Taken together, these results demonstrate that Fnm affects the adherence of E. faecium to fibronectin and is important in the pathogenesis of experimental endocarditis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Endocarditis Bacteriana/microbiología , Enterococcus faecium/patogenicidad , Genoma Bacteriano , Infecciones por Bacterias Grampositivas/microbiología , Proteínas Recombinantes/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Bacteriana , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Sitios de Unión , Colágeno Tipo V/metabolismo , Endocarditis Bacteriana/metabolismo , Endocarditis Bacteriana/patología , Enterococcus faecium/genética , Fibronectinas/metabolismo , Infecciones por Bacterias Grampositivas/metabolismo , Infecciones por Bacterias Grampositivas/patología , Proteínas Inmovilizadas/metabolismo , Laminina/metabolismo , Ratones Endogámicos ICR , Datos de Secuencia Molecular , Unión Proteica , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/metabolismo , Virulencia
14.
J Heart Valve Dis ; 24(2): 236-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26204692

RESUMEN

Libman-Sacks endocarditis is the most widely encountered aseptic endocarditis among patients with systemic lupus erythematosus. Due to the deformed cardiac valves, secondary infective endocarditis should be considered in lupus patients with acute refractory heart failure and fever of unknown origin. The case is reported of a woman with lupus and Libman-Sacks endocarditis who had concurrent coagulase-negative Staphylococcus infective endocarditis that resulted in cerebral septic emboli and acute pulmonary edema. She underwent valve replacement surgery for acute heart failure, and gradually recovered with antibiotic treatment.


Asunto(s)
Endocarditis Bacteriana/complicaciones , Lupus Eritematoso Sistémico/complicaciones , Infecciones Estafilocócicas/complicaciones , Staphylococcus epidermidis , Anciano , Coagulasa/metabolismo , Endocarditis Bacteriana/diagnóstico por imagen , Endocarditis Bacteriana/metabolismo , Endocarditis Bacteriana/microbiología , Femenino , Humanos , Embolia Intracraneal/etiología , Válvula Mitral/diagnóstico por imagen , Válvula Mitral/microbiología , Infecciones Estafilocócicas/metabolismo , Staphylococcus epidermidis/metabolismo , Ultrasonografía
15.
Infect Immun ; 83(6): 2202-12, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25776751

RESUMEN

The mechanisms that underlie valvular inflammation in streptococcus-induced infective endocarditis (IE) remain unclear. We previously demonstrated that streptococcal glucosyltransferases (GTFs) can activate human heart valvular interstitial cells (VIC) to secrete interleukin-6 (IL-6), a cytokine involved in T helper 17 (Th17) cell differentiation. Here, we tested the hypothesis that activated VIC can enhance neutrophil infiltration through sustained IL-17 production, leading to valvular damage. To monitor cytokine and chemokine production, leukocyte recruitment, and the induction or expansion of CD4(+) CD45RA(-) CD25(-) CCR6(+) Th17 cells, primary human VIC were cultured in vitro and activated by GTFs. Serum cytokine levels were measured using an enzyme-linked immunosorbent assay (ELISA), and neutrophils and Th17 cells were detected by immunohistochemistry in infected valves from patients with IE. The expression of IL-21, IL-23, IL-17, and retinoic acid receptor-related orphan receptor C (Rorc) was upregulated in GTF-activated VIC, which may enhance the proliferation of memory Th17 cells in an IL-6-dependent manner. Many chemokines, including chemokine (C-X-C motif) ligand 1 (CXCL1), were upregulated in GTF-activated VIC, which might recruit neutrophils and CD4(+) T cells. Moreover, CXCL1 production in VIC was induced in a dose-dependent manner by IL-17 to enhance neutrophil chemotaxis. CXCL1-expressing VIC and infiltrating neutrophils could be detected in infected valves, and serum concentrations of IL-17, IL-21, and IL-23 were increased in patients with IE compared to healthy donors. Furthermore, elevated serum IL-21 levels have been significantly associated with severe valvular damage, including rupture of chordae tendineae, in IE patients. Our findings suggest that VIC are activated by bacterial modulins to recruit neutrophils and that such activities might be further enhanced by the production of Th17-associated cytokines. Together, these factors can amplify the release of neutrophilic contents in situ, which might lead to severe valvular damage.


Asunto(s)
Endocarditis Bacteriana/metabolismo , Glucosiltransferasas/farmacología , Válvulas Cardíacas/citología , Interleucina-17/metabolismo , Neutrófilos/fisiología , Infecciones Estreptocócicas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/metabolismo , Movimiento Celular , Células Cultivadas , Niño , Preescolar , Femenino , Regulación de la Expresión Génica , Humanos , Inflamación/metabolismo , Interleucina-17/genética , Interleucina-23/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucinas/metabolismo , Masculino , Proteínas de la Membrana , Persona de Mediana Edad , Streptococcus/enzimología , Células Th17/fisiología
16.
J Infect Chemother ; 20(12): 810-3, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25153621

RESUMEN

A 74-year-old man presented with sudden onset of aphasia and apraxia. Magnetic resonance image (MRI) of the brain disclosed a left frontal hemorrhage. The concomitant low grade fever suggestive of infection was unresponsive to cefazolin 1 g q12h, and refractory to piperacillin (PIPC) 2 g q8h. Blood culture grew enterococci, establishing together with echocardiography the diagnosis of infective endocarditis. The angiography revealed cerebral hemorrhage to have resulted from the rupture of the infected intracranial aneurysm. The antimicrobial therapy was switched to ampicillin (ABPC) 2 g q4h plus gentamicin (GM) 60 mg q8h. The positive blood culture was subsequently identified Enterococcus faecium to which the minimum inhibitory concentration (MIC) of PIPC, and ABPC was 16 mcg/mL, and 4 mcg/mL, respectively. The peak concentration of serum ABPC was 83.1, median 50.8, and trough 25.8 mcg/mL. Thus, the percent time > MIC for ABPC was 100%, and the time > minimum bactericidal concentration (MBC) as well. On the other hand, time > MIC for PIPC, was found nearly 30% in retrospective analysis using population pharmacokinetics. The neurological deficit of the patient was completely restored to the normal status after 4-weeks' antimicrobial therapy with ABPC plus GM, then he underwent cardiac surgery for valvular replacement, where microbiological culture of the resected valve was negative. The constellation of the clinical, pharmacological and microbiological outcome in our case provides scientific evidence that the antibiotic therapy given to our case is the best available strategy as an antimicrobial treatment of severe enterococcal endocarditis complicated by disseminated lesion as infected intracranial aneurysm.


Asunto(s)
Aneurisma Roto/microbiología , Antibacterianos/farmacología , Endocarditis Bacteriana/tratamiento farmacológico , Enterococcus faecium/efectos de los fármacos , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Aneurisma Intracraneal/microbiología , Anciano , Ampicilina/farmacocinética , Ampicilina/farmacología , Ampicilina/uso terapéutico , Antibacterianos/farmacocinética , Antibacterianos/uso terapéutico , Encéfalo/patología , Endocarditis Bacteriana/metabolismo , Endocarditis Bacteriana/microbiología , Infecciones por Bacterias Grampositivas/metabolismo , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Imagen por Resonancia Magnética , Masculino , Pruebas de Sensibilidad Microbiana , Resultado del Tratamiento
18.
PLoS Pathog ; 10(1): e1003860, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24453966

RESUMEN

The molecular triggers leading to virulence of a number of human-adapted commensal bacteria such as Streptococcus gallolyticus are largely unknown. This opportunistic pathogen is responsible for endocarditis in the elderly and associated with colorectal cancer. Colonization of damaged host tissues with exposed collagen, such as cardiac valves and pre-cancerous polyps, is mediated by appendages referred to as Pil1 pili. Populations of S. gallolyticus are heterogeneous with the majority of cells weakly piliated while a smaller fraction is hyper piliated. We provide genetic evidences that heterogeneous pil1 expression depends on a phase variation mechanism involving addition/deletion of GCAGA repeats that modifies the length of an upstream leader peptide. Synthesis of longer leader peptides potentiates the transcription of the pil1 genes through ribosome-induced destabilization of a premature stem-loop transcription terminator. This study describes, at the molecular level, a new regulatory mechanism combining phase variation in a leader peptide-encoding gene and transcription attenuation. This simple and robust mechanism controls a stochastic heterogeneous pilus expression, which is important for evading the host immune system while ensuring optimal tissue colonization.


Asunto(s)
Proteínas Fimbrias/biosíntesis , Fimbrias Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Streptococcus/metabolismo , Endocarditis Bacteriana/genética , Endocarditis Bacteriana/metabolismo , Proteínas Fimbrias/genética , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/ultraestructura , Humanos , Procesos Estocásticos , Streptococcus/genética , Streptococcus/ultraestructura
19.
Circulation ; 128(1): 50-9, 2013 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-23720451

RESUMEN

BACKGROUND: During pathogenesis of infective endocarditis, Staphylococcus aureus adherence often occurs without identifiable preexisting heart disease. However, molecular mechanisms mediating initial bacterial adhesion to morphologically intact endocardium are largely unknown. METHODS AND RESULTS: Perfusion of activated human endothelial cells with fluorescent bacteria under high-shear-rate conditions revealed 95% attachment of the S aureus by ultralarge von Willebrand factor (ULVWF). Flow experiments with VWF deletion mutants and heparin indicate a contribution of the A-type domains of VWF to bacterial binding. In this context, analyses of different bacterial deletion mutants suggest the involvement of wall teichoic acid but not of staphylococcal protein A. The presence of inactivated platelets and serum increased significantly ULVWF-mediated bacterial adherence. ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin motifs 13) caused a dose-dependent reduction of bacterial binding and a reduced length of ULVWF, but single cocci were still tethered by ULVWF at physiological levels of ADAMTS13. To further prove the role of VWF in vivo, we compared wild-type mice with VWF knockout mice. Binding of fluorescent bacteria was followed in tumor necrosis factor-α-stimulated tissue by intravital microscopy applying the dorsal skinfold chamber model. Compared with wild-type mice (n=6), we found less bacteria in postcapillary (60±6 versus 32±5 bacteria) and collecting venules (48±5 versus 18±4 bacteria; P<0.05) of VWF knockout mice (n=5). CONCLUSIONS: Our data provide the first evidence that ULVWF contributes to the initial pathogenic step of S aureus-induced endocarditis in patients with an apparently intact endothelium. An intervention reducing the ULVWF formation with heparin or ADAMTS13 suggests novel therapeutic options to prevent infective endocarditis.


Asunto(s)
Endocarditis Bacteriana/metabolismo , Células Endoteliales/microbiología , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/metabolismo , Factor de von Willebrand/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAMTS13 , Animales , Anticoagulantes/metabolismo , Anticoagulantes/farmacología , Adhesión Bacteriana/fisiología , Plaquetas/metabolismo , Plaquetas/microbiología , Endocarditis Bacteriana/microbiología , Endocarditis Bacteriana/prevención & control , Células Endoteliales/citología , Células Endoteliales/fisiología , Fibrinógeno/metabolismo , Fibrinógeno/farmacología , Heparina/metabolismo , Heparina/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Metaloendopeptidasas/metabolismo , Ratones , Ratones Noqueados , Tamaño de la Partícula , Piel/citología , Piel/microbiología , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus/patogenicidad , Estrés Mecánico , Factores de Virulencia/metabolismo , Factor de von Willebrand/química , Factor de von Willebrand/genética
20.
Oral Dis ; 19(4): 387-93, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22998492

RESUMEN

OBJECTIVE: Streptococcus mutans, a major pathogen of dental caries, is considered to be one of the causative agents of infective endocarditis (IE). Two types of cell surface collagen-binding proteins, Cnm and Cbm, have been identified in the organism. The aim of the present study was to analyze these proteins as possible etiologic factors for IE. MATERIALS AND METHODS: The binding activities of S. mutans strains to collagen types I, III, and IV were analyzed relative to the presence of Cnm and Cbm, as were their adhesion and invasion properties with human umbilical vein endothelial cells (HUVEC). In addition, distributions of the genes encoding Cnm and Cbm in S. mutans-positive heart valve specimens extirpated from IE and non-IE patients were analyzed by PCR. RESULTS: Most of the Cbm-positive strains showed higher levels of binding to type I collagen as well as higher rates of adhesion and invasion with HUVEC as compared to the Cnm-positive strains. Furthermore, the gene encoding Cbm was detected significantly more frequently in heart valve specimens from IE patients than from non-IE patients. CONCLUSIONS: These results suggest that the collagen-binding protein Cbm of S. mutans may be one of the potential important factor associated with the pathogenesis of IE.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Colágeno/metabolismo , Endocarditis Bacteriana/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus mutans/química , Adhesinas Bacterianas/genética , Adulto , Anciano , Válvula Aórtica/microbiología , Adhesión Bacteriana , Proteínas Portadoras/genética , ADN Bacteriano , Endocarditis Bacteriana/metabolismo , Células Endoteliales/microbiología , Femenino , Células Endoteliales de la Vena Umbilical Humana/microbiología , Humanos , Masculino , Persona de Mediana Edad , Válvula Mitral/microbiología , Unión Proteica , Infecciones Estreptocócicas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...