Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34725157

RESUMEN

Neisseria meningitidis utilizes type IV pili (T4P) to adhere to and colonize host endothelial cells, a process at the heart of meningococcal invasive diseases leading to meningitis and sepsis. T4P are polymers of an antigenically variable major pilin building block, PilE, plus several core minor pilins that initiate pilus assembly and are thought to be located at the pilus tip. Adhesion of N. meningitidis to human endothelial cells requires both PilE and a conserved noncore minor pilin PilV, but the localization of PilV and its precise role in this process remains to be clarified. Here, we show that both PilE and PilV promote adhesion to endothelial vessels in vivo. The substantial adhesion defect observed for pilV mutants suggests it is the main adhesin. Consistent with this observation, superresolution microscopy showed the abundant distribution of PilV throughout the pilus. We determined the crystal structure of PilV and modeled it within the pilus filament. The small size of PilV causes it to be recessed relative to adjacent PilE subunits, which are dominated by a prominent hypervariable loop. Nonetheless, we identified a conserved surface-exposed adhesive loop on PilV by alanine scanning mutagenesis. Critically, antibodies directed against PilV inhibit N. meningitidis colonization of human skin grafts. These findings explain how N. meningitidis T4P undergo antigenic variation to evade the humoral immune response while maintaining their adhesive function and establish the potential of this highly conserved minor pilin as a vaccine and therapeutic target for the prevention and treatment of N. meningitidis infections.


Asunto(s)
Adhesión Bacteriana , Proteínas Bacterianas/fisiología , Fimbrias Bacterianas/fisiología , Neisseria meningitidis/fisiología , Animales , Anticuerpos/uso terapéutico , Proteínas Bacterianas/química , Proteínas Bacterianas/ultraestructura , Línea Celular , Evaluación Preclínica de Medicamentos , Femenino , Fimbrias Bacterianas/química , Fimbrias Bacterianas/ultraestructura , Humanos , Infecciones Meningocócicas/tratamiento farmacológico , Ratones SCID
2.
Proc Natl Acad Sci U S A ; 117(5): 2606-2612, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31964828

RESUMEN

Bacterial infections are frequently based on the binding of lectin-like adhesins to specific glycan determinants exposed on host cell receptors. These interactions confer species-specific recognition and tropism for particular host tissues and represent attractive antibacterial targets. However, the wide structural diversity of carbohydrates hampers the characterization of specific glycan determinants. Here, we characterized the receptor recognition of type IV pili (Tfp), a key adhesive factor present in numerous bacterial pathogens, using Neisseria meningitidis as a model organism. We found that meningococcal Tfp specifically recognize a triantennary sialylated poly-N-acetyllactosamine-containing N-glycan exposed on the human receptor CD147/Basigin, while fucosylated derivatives of this N-glycan impaired bacterial adhesion. Corroborating the inhibitory role of fucosylation on receptor recognition, adhesion of the meningococcus on nonhuman cells expressing human CD147 required prior defucosylation. These findings reveal the molecular basis of the selective receptor recognition by meningococcal Tfp and thereby, identify a potential antibacterial target.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Proteínas Fimbrias/metabolismo , Infecciones Meningocócicas/metabolismo , Neisseria meningitidis/metabolismo , Receptores de Superficie Celular/metabolismo , Adhesinas Bacterianas/genética , Proteínas Fimbrias/genética , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/metabolismo , Glicosilación , Humanos , Infecciones Meningocócicas/genética , Infecciones Meningocócicas/microbiología , Neisseria meningitidis/genética , Polisacáridos/metabolismo , Receptores de Superficie Celular/genética
3.
Nat Commun ; 10(1): 4752, 2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31628314

RESUMEN

Meningococcus utilizes ß-arrestin selective activation of endothelial cell ß2 adrenergic receptor (ß2AR) to cause meningitis in humans. Molecular mechanisms of receptor activation by the pathogen and of its species selectivity remained elusive. We report that ß2AR activation requires two asparagine-branched glycan chains with terminally exposed N-acetyl-neuraminic acid (sialic acid, Neu5Ac) residues located at a specific distance in its N-terminus, while being independent of surrounding amino-acid residues. Meningococcus triggers receptor signaling by exerting direct and hemodynamic-promoted traction forces on ß2AR glycans. Similar activation is recapitulated with beads coated with Neu5Ac-binding lectins, submitted to mechanical stimulation. This previously unknown glycan-dependent mode of allosteric mechanical activation of a G protein-coupled receptor contributes to meningococcal species selectivity, since Neu5Ac is only abundant in humans due to the loss of CMAH, the enzyme converting Neu5Ac into N-glycolyl-neuraminic acid in other mammals. It represents an additional mechanism of evolutionary adaptation of a pathogen to its host.


Asunto(s)
Fimbrias Bacterianas/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Neisseria meningitidis/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Línea Celular , Membrana Celular/metabolismo , Fimbrias Bacterianas/genética , Células HEK293 , Humanos , Lectinas/metabolismo , Microscopía Confocal , Neisseria meningitidis/fisiología , Polisacáridos/metabolismo , Receptores Adrenérgicos beta 2/genética , Homología de Secuencia de Aminoácido , beta-Arrestinas/metabolismo
4.
Methods Mol Biol ; 1957: 325-334, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30919363

RESUMEN

Neisseria meningitidis is a Gram-negative diplococcus restricted to humans that causes severe septicemia and/or meningitidis. Initial adhesion to human endothelial cells is mediated through the interaction of type IV pili with the hetero-oligomeric complexes formed by the human receptors CD147 and the ß2-adrenergic receptor. Interaction with this complex heterodimer activates a ß-arrestin-biased signaling pathway leading to actin polymerization and accumulation of ezrin and ezrin-binding partners. These signaling events promote the formation of cell plasma membrane protrusions in endothelial cells, which are crucial for N. meningitidis colonies to resist shear stress and colonize blood vessels. Here we provide detailed protocols to evaluate the role of ß-arrestins in actin and ezrin signaling downstream of G protein-coupled receptor activation.


Asunto(s)
Biología Molecular/métodos , Neisseria meningitidis/metabolismo , Transducción de Señal , beta-Arrestinas/metabolismo , Células Cultivadas , Células Endoteliales/metabolismo , Células Endoteliales/microbiología , Células HEK293 , Humanos , ARN Interferente Pequeño/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
5.
PLoS Pathog ; 14(4): e1006981, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29630665

RESUMEN

Purpura fulminans is a deadly complication of Neisseria meningitidis infections due to extensive thrombosis of microvessels. Although a Disseminated Intra-vascular Coagulation syndrome (DIC) is frequently observed during Gram negative sepsis, it is rarely associated with extensive thrombosis like those observed during meningococcemia, suggesting that the meningococcus induces a specific dysregulation of coagulation. Another specific feature of N. meningitidis pathogenesis is its ability to colonize microvessels endothelial cells via type IV pili. Importantly, endothelial cells are key in controlling the coagulation cascade through the activation of the potent anticoagulant Protein C (PC) thanks to two endothelial cell receptors among which the Endothelial Protein C Receptor (EPCR). Considering that congenital or acquired deficiencies of PC are associated with purpura fulminans, we hypothesized that a defect in the activation of PC following meningococcal adhesion to microvessels is responsible for the thrombotic events observed during meningococcemia. Here we showed that the adhesion of N. meningitidis on endothelial cells results in a rapid and intense decrease of EPCR expression by inducing its cleavage in a process know as shedding. Using siRNA experiments and CRISPR/Cas9 genome edition we identified ADAM10 (A Disintegrin And Metalloproteinase-10) as the protease responsible for this shedding. Surprisingly, ADAM17, the only EPCR sheddase described so far, was not involved in this process. Finally, we showed that this ADAM10-mediated shedding of EPCR induced by the meningococcal interaction with endothelial cells was responsible for an impaired activation of Protein C. This work unveils for the first time a direct link between meningococcal adhesion to endothelial cells and a severe dysregulation of coagulation, and potentially identifies new therapeutic targets for meningococcal purpura fulminans.


Asunto(s)
Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Endotelio Vascular/patología , Proteínas de la Membrana/metabolismo , Infecciones Meningocócicas/complicaciones , Microvasos/patología , Proteína C/metabolismo , Púrpura Fulminante/etiología , Proteína ADAM10/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Adhesión Bacteriana , Coagulación Sanguínea/fisiología , Células Cultivadas , Receptor de Proteína C Endotelial/genética , Endotelio Vascular/metabolismo , Endotelio Vascular/microbiología , Humanos , Proteínas de la Membrana/genética , Infecciones Meningocócicas/microbiología , Microvasos/metabolismo , Microvasos/microbiología , Neisseria meningitidis/fisiología , Proteína C/genética , Púrpura Fulminante/metabolismo , Púrpura Fulminante/patología
6.
Virulence ; 8(8): 1808-1819, 2017 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-29099305

RESUMEN

Neisseria meningitidis is the causative agent of cerebrospinal meningitis and that of a rapidly progressing fatal septic shock known as purpura fulminans. Meningococcemia is characterized by bacterial adhesion to human endothelial cells of the microvessels. Host specificity has hampered studies on the role of blood vessels colonization in N. meningitidis associated pathogenesis. In this work, using a humanized model of SCID mice allowing the study of bacterial adhesion to human cells in an in vivo context we demonstrate that meningococcal colonization of human blood vessels is a prerequisite to the establishment of sepsis and lethality. To identify the molecular pathways involved in bacterial virulence, we performed transposon insertion site sequencing (Tn-seq) in vivo. Our results demonstrate that 36% of the genes that are important for growth in the blood of mice are dispensable when bacteria colonize human blood vessels, suggesting that human endothelial cells lining the blood vessels are feeding niches for N. meningitidis in vivo. Altogether, our work proposes a new paradigm for meningococcal virulence in which colonization of blood vessels is associated with metabolic adaptation and sustained bacteremia responsible for sepsis and subsequent lethality.


Asunto(s)
Bacteriemia/microbiología , Infecciones Meningocócicas/sangre , Infecciones Meningocócicas/microbiología , Microvasos/microbiología , Neisseria meningitidis/fisiología , Animales , Bacteriemia/sangre , Adhesión Bacteriana , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Neisseria meningitidis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...