Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Front Microbiol ; 10: 742, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31105650

RESUMEN

Until 2007, Staphylococcus aureus from clonal complex 398 (CC398) was exclusively associated with livestock species and companion animals. Recently, several studies described the emergence of S. aureus CC398 as etiologies of severe infections in humans living in an animal-free environment. Recent sequencing efforts showed that the mobile genetic elements found in CC398 isolates were specific for each population and enabled differentiation of strains responsible for asymptomatic colonization from strains involved in bloodstream infections. We mobilized prophages from a human CC398 isolate and introduced them into two naïve ancestral isolates devoid of prophages that exclusively colonize animals. These lysogenized ancestral CC398 isolates acquired features related to virulence, such as an increased capacity to adhere to human extracellular matrix proteins and the ability to invade and survive within non-phagocytic cells. Pathogenicity of several clinical isolates from the CC398 lineage as well as ancestral and in vitro lysogenized ancestral counterparts was assessed in a model of infectious endocarditis in rats. Natural and artificial lysogens were not only more invasive than their prophage-free parent but also showed an increased capacity to multiply within aortic vegetations. This study identified prophages as mediators of bacterial virulence in a model of infectious endocarditis, probably through promotion of interaction with extracellular matrix components. Further studies are needed to identify mechanisms leading to promotion of intrinsic virulence.

2.
Virulence ; 9(1): 1615-1624, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30280967

RESUMEN

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.


Asunto(s)
Válvula Aórtica/microbiología , Proteínas Bacterianas/metabolismo , Coagulasa/metabolismo , Endocarditis Bacteriana/patología , Staphylococcus aureus/genética , Factor de von Willebrand/metabolismo , Animales , Válvula Aórtica/fisiopatología , Proteínas Bacterianas/genética , Infecciones Relacionadas con Catéteres/microbiología , Coagulasa/genética , Femenino , Eliminación de Gen , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Ratas , Ratas Wistar , Infecciones Estafilocócicas , Staphylococcus aureus/patogenicidad , Factores de Virulencia/genética
3.
Water Res ; 143: 334-345, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-29986243

RESUMEN

The emergence of antibiotic resistance represents a major threat to human health. In this work we investigated the elimination of antibiotic resistant bacteria (ARB) by solar light and solar photo-Fenton processes. As such, we have designed an experimental plan in which several bacterial strains (Staphylococcus aureus, Escherichia coli and Klebsiella pneumoniae) possessing different drug-susceptible and -resistant patterns and structures (Gram-positive and Gram-negative) were subjected to solar light and the photo-Fenton oxidative treatment in water. We showed that both solar light and solar photo-Fenton processes were effective in the elimination of ARB in water and that the time necessary for solar light disinfection and solar photo-Fenton disinfection were similar for antibiotic-susceptible and antibiotic-resistant strains (mostly 180-240 and 90-120 min, respectively). Moreover, the bacterial structure did not significantly affect the effectiveness of the treatment. Similar regrowth pattern was observed (compared to the susceptible strain) and no development of bacteria with higher drug-resistance values was found in waters after any treatment. Finally, both processes were effective to reduce AR genes (ARGs), although solar photo-Fenton was more rapid than solar light. In conclusion, the solar photo-Fenton process ensured effective disinfection of ARB and elimination of ARGs in water (or wastewater) and is a potential mean to ensure limitation of ARB and ARG spread in nature.


Asunto(s)
Antibacterianos/farmacología , Bacterias/genética , Desinfección/métodos , Farmacorresistencia Bacteriana/genética , Bacterias/efectos de los fármacos , Farmacorresistencia Bacteriana/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Oxidación-Reducción , Fotoquímica/métodos , Luz Solar , Aguas Residuales/microbiología , Microbiología del Agua , Purificación del Agua/métodos
4.
PLoS One ; 13(2): e0192507, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29408864

RESUMEN

The recent rise of multidrug-resistant Gram-negative bacteria represents a serious threat to public health and makes the search for novel effective alternatives to antibiotics a compelling need. Bacteriophage (Phage) lysins are enzymes that hydrolyze the cell wall of bacteria and represent a promising alternative to tackle this ever-increasing problem. Despite their use is believed to be restricted to Gram-positive bacteria, recent findings have shown that they can also be used against Gram-negative bacteria. By using a phage genome-based screening approach, we identified and characterized a novel lysin, PlyE146, encoded by an Escherichia coli prophage and with a predicted molecular mass of ca. 17 kDa. PlyE146 is composed of a C-terminal cationic peptide and a N-terminal N-acetylmuramidase domain. Histidine-tagged PlyE146 was overexpressed from a plasmid in Lactococcus lactis NZ9000 and purified by NI-NTA chromatography. PlyE146 exhibited in vitro optimal bactericidal activity against E. coli K12 (3.6 log10 CFU/mL decrease) after 2 h of incubation at 37°C at a concentration of 400 µg/mL in the absence of NaCl and at pH 6.0. Under these conditions, PlyE146 displayed antimicrobial activity towards several other E. coli, Pseudomonas aeruginosa (3 to 3.8-log10 CFU/mL decrease) and Acinetobacter baumannii (4.9 to >5-log10 CFU/mL decrease) strains. Therefore, PlyE146 represents a promising therapeutic agent against E. coli, P. aeruginosa and A. baumannii infections. However, further studies are required to improve the efficacy of PlyE146 under physiological conditions.


Asunto(s)
Colifagos/metabolismo , Bacterias Gramnegativas/metabolismo , Antibacterianos/farmacología , Western Blotting , Glicósido Hidrolasas/metabolismo , Bacterias Gramnegativas/efectos de los fármacos , Microscopía Electrónica de Transmisión
5.
Infect Immun ; 84(12): 3557-3563, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27736784

RESUMEN

Streptococcus gordonii and related species of oral viridans group streptococci (VGS) are common etiological agents of infective endocarditis (IE). We explored vaccination as a strategy to prevent VGS-IE, using a novel antigen-presenting system based on non-genetically modified Lactococcus lactis displaying vaccinogens on its surface. Hsa and PadA are surface-located S. gordonii proteins implicated in platelet adhesion and aggregation, which are key steps in the pathogenesis of IE. This function makes them ideal targets for vaccination against VGS-IE. In the present study, we report the use of nonliving L. lactis displaying at its surface the N-terminal region of Hsa or PadA by means of the cell wall binding domain of Lactobacillus casei A2 phage lysine LysA2 (Hsa-LysA2 and PadA-LysA2, respectively) and investigation of their ability to elicit antibodies in rats and to protect them from S. gordonii experimental IE. Immunized and control animals with catheter-induced sterile aortic valve vegetations were inoculated with 106 CFU of S. gordonii The presence of IE was evaluated 24 h later. Immunization of rats with L. lactis Hsa-LysA2, L. lactis PadA-LysA2, or both protected 6/11 (55%), 6/11 (55%), and 11/12 (91%) animals, respectively, from S. gordonii IE (P < 0.05 versus controls). Protection correlated with the induction of high levels of functional antibodies against both Hsa and PadA that delayed or totally inhibited platelet aggregation by S. gordonii These results support the value of L. lactis as a system for antigen delivery and of Hsa and PadA as promising candidates for a vaccine against VGS-IE.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Anticuerpos Antibacterianos/inmunología , Proteínas Portadoras/metabolismo , Endocarditis Bacteriana/prevención & control , Agregación Plaquetaria/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus gordonii/metabolismo , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/inmunología , Animales , Vacunas Bacterianas/inmunología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Femenino , Regulación Bacteriana de la Expresión Génica , Hemaglutininas Virales , Lactobacillus leichmannii/genética , Lactobacillus leichmannii/metabolismo , Ratas
6.
PLoS One ; 11(8): e0160554, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27505001

RESUMEN

Streptococcus tigurinus is responsible for severe invasive infections such as infective endocarditis, spondylodiscitis and meningitis. As described, S. tigurinus isolates AZ_3aT and AZ_14 were highly virulent (HV phenotype) in an experimental model of infective endocarditis and showed enhanced adherence and invasion of human endothelial cells when compared to low virulent S. tigurinus isolate AZ_8 (LV phenotype). Here, we sought whether genetic determinants could explain the higher virulence of AZ_3aT and AZ_14 isolates. Several genetic determinants specific to the HV strains were identified through extensive comparative genomics amongst which some were thought to be highly relevant for the observed HV phenotype. These included i) an iron uptake and metabolism operon, ii) an ascorbate assimilation operon, iii) a newly acquired PI-2-like pilus islets described for the first time in S. tigurinus, iv) a hyaluronate metabolism operon, v) an Entner-Doudoroff pathway of carbohydrates metabolism, and vi) an alternate pathways for indole biosynthesis. We believe that the identified genomic features could largely explain the phenotype of high infectivity of the two HV S. tigurinus strains. Indeed, these features include determinants that could be involved at different stages of the disease such as survival of S. tigurinus in blood (iron uptake and ascorbate metabolism operons), initial attachment of bacterial pathogen to the damaged cardiac tissue and/or vegetation that formed on site (PI-2-like pilus islets), tissue invasion (hyaluronate operon and Entner-Doudoroff pathway) and regulation of pathogenicity (indole biosynthesis pathway).


Asunto(s)
Genómica , Streptococcus/genética , Streptococcus/patogenicidad , Ácido Ascórbico/metabolismo , Adhesión Bacteriana/genética , Transporte Biológico/genética , Ácido Hialurónico/metabolismo , Indoles/metabolismo , Hierro/metabolismo , Anotación de Secuencia Molecular , Fenotipo , Proteómica , Especificidad de la Especie , Streptococcus/metabolismo , Triptófano/metabolismo , Virulencia/genética
7.
ACS Med Chem Lett ; 7(6): 606-11, 2016 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-27326335

RESUMEN

Sortases are transpeptidase enzymes that anchor surface proteins, including virulence factors, to the cell wall of Gram-positive bacteria, and they are potential targets for the development of anti-infective agents. While several large compound libraries were searched by high-throughput screening, no high-affinity inhibitors of sortases could be developed to date. Here, we applied phage display to screen billions of peptide macrocycles against sortase A (SrtA) of Staphylococcus aureus (S. aureus). We were able to identify potent and selective inhibitors of SrtA that blocked SrtA-mediated anchoring of synthetic substrates to the surface of live S. aureus cells. A region present in all inhibitory peptides (Leu-Pro-Pro) resembled the natural substrates of SrtA (Leu-Pro-Xaa-Thr-Gly), suggesting that the macrocycles bind to the enzyme's active site and that they form similar molecular contacts as natural substrates. The evolved peptide macrocycles may be used as lead structures for the development of potent peptidomimetic SrtA inhibitors.

8.
Pathog Dis ; 73(8): ftv060, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26310867

RESUMEN

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.


Asunto(s)
Aspirina/administración & dosificación , Endocarditis Bacteriana/prevención & control , Enterococcus faecalis/crecimiento & desarrollo , Infecciones por Bacterias Grampositivas/prevención & control , Inhibidores de Agregación Plaquetaria/farmacología , Streptococcus/crecimiento & desarrollo , Ticlopidina/administración & dosificación , Animales , Modelos Animales de Enfermedad , Endocarditis Bacteriana/microbiología , Femenino , Infecciones por Bacterias Grampositivas/microbiología , Ratas Wistar , Resultado del Tratamiento
9.
Vaccine ; 33(30): 3512-7, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26048778

RESUMEN

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.


Asunto(s)
Adhesinas Bacterianas/inmunología , Coagulasa/inmunología , Portadores de Fármacos , Endocarditis/prevención & control , Lactococcus lactis/genética , Vacunas Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Adhesinas Bacterianas/genética , Animales , Anticuerpos Antibacterianos/sangre , Coagulasa/genética , Modelos Animales de Enfermedad , Endocarditis/inmunología , Femenino , Fibronectinas/metabolismo , Ratas Wistar , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Vacunas Estafilocócicas/administración & dosificación , Vacunas Estafilocócicas/genética , Staphylococcus aureus/genética , Resultado del Tratamiento , Vacunación/métodos
11.
J Infect Dis ; 211(1): 72-9, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25086177

RESUMEN

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.


Asunto(s)
Endocarditis Bacteriana/tratamiento farmacológico , Fibrinolíticos/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Animales , Profilaxis Antibiótica/métodos , Bacteriemia/tratamiento farmacológico , Bacteriemia/microbiología , Modelos Animales de Enfermedad , Endocarditis Bacteriana/microbiología , Humanos , Ratas , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/efectos de los fármacos , Streptococcus gordonii/efectos de los fármacos
12.
Int J Med Microbiol ; 303(8): 498-504, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23856340

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Endocarditis Bacteriana/microbiología , Endocarditis Bacteriana/patología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/patología , Streptococcus/patogenicidad , Animales , Aorta/microbiología , Aorta/patología , Células Endoteliales/microbiología , Macrófagos/inmunología , Macrófagos/microbiología , Fagocitosis , Ratas , Streptococcus/inmunología
13.
Infect Immun ; 81(3): 697-703, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23250949

RESUMEN

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.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Bacteriemia/inmunología , Endocarditis Bacteriana/inmunología , Agregación Plaquetaria/inmunología , Staphylococcus aureus/metabolismo , Adhesinas Bacterianas/genética , Animales , Adhesión Bacteriana/fisiología , Citocinas/genética , Citocinas/metabolismo , Fibrinógeno , Fibronectinas , Regulación de la Expresión Génica/inmunología , Humanos , Proteínas Inmovilizadas , Ratas
14.
Infect Immun ; 78(12): 5195-201, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20837714

RESUMEN

Parachlamydia acanthamoebae is a Chlamydia-related organism whose pathogenic role in pneumonia is supported by serological and molecular clinical studies and an experimental mouse model of lung infection. Toll-like receptors (TLRs) play a seminal role in sensing microbial products and initiating innate immune responses. The aim of this study was to investigate the roles of MyD88, TLR2, and TLR4 in the interaction of Parachlamydia with macrophages. Here, we showed that Parachlamydia entered bone-marrow derived macrophages (BMDMs) in a TLR-independent manner but did not multiply intracellularly. Interestingly, compared to live bacteria, heat-inactivated Parachlamydia induced the production of substantial amounts of tumor necrosis factor alpha (TNF), interleukin-6 (IL-6), and IL-12p40 by BMDMs and of TNF and IL-6 by peritoneal macrophages as well as RAW 264.7 and J774 macrophage cell lines. Cytokine production by BMDMs, which was partially inhibited upon trypsin treatment of Parachlamydia, was dependent on MyD88, TLR4, and, to a lesser extent, TLR2. Finally, MyD88(-/-), TLR4(-/-), and TLR2(-/-) mice were as resistant as wild-type mice to lung infection following the intratracheal instillation of Parachlamydia. Thus, in contrast to Chlamydia pneumoniae, Parachlamydia acanthamoebae weakly stimulates macrophages, potentially compensating for its low replication capacity in macrophages by escaping the innate immune surveillance.


Asunto(s)
Chlamydiales/fisiología , Factor 88 de Diferenciación Mieloide/fisiología , Receptor Toll-Like 2/fisiología , Receptor Toll-Like 4/fisiología , Animales , Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/microbiología , Infecciones por Chlamydia/patología , Chlamydiales/inmunología , Femenino , Interacciones Huésped-Patógeno/inmunología , Interacciones Huésped-Patógeno/fisiología , Interleucina-6/análisis , Interleucina-6/sangre , Interleucina-6/fisiología , Pulmón/química , Pulmón/inmunología , Pulmón/patología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/inmunología , Fagocitosis/fisiología , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología , Transducción de Señal/fisiología , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/inmunología , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/fisiología
15.
Microb Pathog ; 45(2): 92-7, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18502092

RESUMEN

The role of Parachlamydia acanthamoebae as an agent of pneumonia is suggested by sero-epidemiological studies, molecular surveys and by the permissivity of macrophages, lung fibroblasts and pneumocytes to this obligate intracellular bacteria. We thus developed a murine model of pneumonia due to Parachlamydia. Mice were inoculated intratracheally with Parachlamydia acanthamoebae. Pneumonia-associated mortality was of 50% 5 days post-inoculation. Lungs histopathology was characterized by purulent and interstitial pneumonia. The presence of Parachlamydia in the lesions was demonstrated by PCR, immunohistochemistry and electron microscopy. Moreover, living Parachlamydia could be recovered from the lungs of infected mice using amoebal co-culture. All control mice inoculated with heat-inactivated bacteria were free of symptoms and survived. Thus, we demonstrated that Parachlamydia induce a severe pneumonia in mice. This animal model, which confirms the third and fourth Koch postulates, may be suitable to test in vivo efficient therapeutic regimens against Parachlamydia.


Asunto(s)
Chlamydiales/patogenicidad , Pulmón/patología , Neumonía/microbiología , Animales , Chlamydiales/fisiología , Modelos Animales de Enfermedad , Pulmón/microbiología , Ratones , Microscopía Electrónica , Modelos Biológicos , Neumonía/fisiopatología
16.
Infect Immun ; 73(2): 990-8, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15664942

RESUMEN

Isogenic Staphylococcus aureus strains with different capacities to produce sigma(B) activity were analyzed for their ability to attach to fibrinogen- or fibronectin-coated surfaces or platelet-fibrin clots and to cause endocarditis in rats. In comparison to the sigma(B)-deficient strain, BB255, which harbors an rsbU mutation, both rsbU-complemented and sigma(B)-overproducing derivatives exhibited at least five times greater attachment to fibrinogen- and fibronectin-coated surfaces and showed increased adherence to platelet-fibrin clots. No differences in adherence were seen between BB255 and a DeltarsbUVWsigB isogen. Northern blotting analyses revealed that transcription of clfA, encoding fibrinogen-binding protein clumping factor A, and fnbA, encoding fibronectin-binding protein A, were positively influenced by sigma(B). Sigma(B) overproduction resulted in a statistically significant increase in positive spleen cultures and enhanced bacterial densities in both the aortic vegetations and spleens at 16 h postinoculation. In contrast, at 72 h postinoculation, tissues infected with the sigma(B) overproducer had lower bacterial densities than did those infected with BB255. These results suggest that although sigma(B) appears to increase the adhesion of S. aureus to various host cell-matrix proteins in vitro, it has limited effect on pathogenesis in the rat endocarditis model. Sigma(B) appears to have a transient enhancing effect on bacterial density in the early stages of infection that is lost during progression.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Proteínas Bacterianas/metabolismo , Endocarditis Bacteriana/metabolismo , Factor sigma/metabolismo , Staphylococcus aureus/metabolismo , Adhesinas Bacterianas/genética , Animales , Adhesión Celular/fisiología , Pared Celular , Endocarditis Bacteriana/microbiología , Fibrinógeno/metabolismo , Fibronectinas/metabolismo , Regiones Promotoras Genéticas , Ratas , Staphylococcus aureus/patogenicidad , alfa-Macroglobulinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA