Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nano Lett ; 24(28): 8567-8574, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-38959438

RESUMEN

Phagocytosis is an essential mechanism of the human immune system where pathogens are eliminated by immune cells. The CCN1 protein plays an important role in the phagocytosis of Staphylococcus aureus by favoring the bridging of the αVß3 integrin to the bacterial peptidoglycan (PG), through mechanical forces that remain unknown. Here, we employ single-molecule experiments to unravel the nanomechanics of the PG-CCN1-αVß3 ternary complex. While CCN1 binds αVß3 integrins with moderate force (∼60 pN), much higher binding strengths (up to ∼800 pN) are observed between CCN1 and PG. Notably, the strength of both CCN1-αVß3 and CCN1-PG bonds is dramatically enhanced by tensile loading, favoring a model in which mechanical stress induces the exposure of cryptic integrin binding sites in CCN1 and multivalent binding between CCN1 lectin sites and monosaccharides along the PG glycan chains.


Asunto(s)
Proteína 61 Rica en Cisteína , Integrina alfaVbeta3 , Fagocitosis , Staphylococcus aureus , Staphylococcus aureus/metabolismo , Staphylococcus aureus/fisiología , Humanos , Proteína 61 Rica en Cisteína/metabolismo , Proteína 61 Rica en Cisteína/química , Integrina alfaVbeta3/metabolismo , Peptidoglicano/metabolismo , Peptidoglicano/química , Unión Proteica , Sitios de Unión
2.
Nano Lett ; 23(11): 5297-5306, 2023 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-37267288

RESUMEN

Various viruses and pathogenic bacteria interact with annexin A2 to invade mammalian cells. Here, we show that Staphylococcus aureus engages in extremely strong catch bonds for host cell invasion. By means of single-molecule atomic force microscopy, we find that bacterial surface-located clumping factors bind annexin A2 with extraordinary strength, indicating that these bonds are extremely resilient to mechanical tension. By determining the lifetimes of the complexes under increasing mechanical stress, we demonstrate that the adhesins form catch bonds with their ligand that are capable to sustain forces of 1500-1700 pN. The force-dependent adhesion mechanism identified here provides a molecular framework to explain how S. aureus pathogens tightly attach to host cells during invasion and shows promise for the design of new therapeutics against intracellular S. aureus.


Asunto(s)
Anexina A2 , Staphylococcus aureus , Adhesión Bacteriana , Anexina A2/metabolismo , Unión Proteica , Adhesinas Bacterianas/química
3.
Mol Microbiol ; 117(6): 1479-1492, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35570359

RESUMEN

The binding of Streptococcus pneumoniae to collagen is likely an important step in the pathogenesis of pneumococcal infections, but little is known of the underlying molecular mechanisms. Streptococcal surface repeats (SSURE) are highly conserved protein domains present in cell wall adhesins from different Streptococcus species. We find here that SSURE repeats of the pneumococcal adhesin plasminogen and fibronectin binding protein B (PfbB) bind to various types of collagen. Moreover, deletion of the pfbB gene resulted in a significant impairment of the ability of encapsulated or unencapsulated pneumococci to bind collagen. Notably, a PfbB SSURE domain is also bound to the complement component C1q that bears a collagen-like domain and promotes adherence of pneumococci to host cells by acting as a bridge between bacteria and epithelial cells. Accordingly, deletion of PfbB or pre-treatment with anti-SSURE antibodies markedly decreased pneumococcal binding to C1q as well as C1q-dependent adherence to epithelial and endothelial cells. Further data indicated that C1q promotes pneumococcal adherence by binding to integrin α2 ß1 . In conclusion, our results indicate that the SSURE domains of the PfbB protein promote interactions of pneumococci with various types of collagen and with C1q. These repeats may be useful targets in strategies to control S. pneumoniae infections.


Asunto(s)
Infecciones Neumocócicas , Streptococcus pneumoniae , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Colágeno/genética , Colágeno/metabolismo , Complemento C1q/genética , Complemento C1q/metabolismo , Células Endoteliales/metabolismo , Humanos , Infecciones Neumocócicas/metabolismo , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/metabolismo
4.
Int J Mol Sci ; 24(21)2023 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-37958706

RESUMEN

Staphylococcus lugdunensis is an emerging high-virulent pathogen. Here, the presence and expression of virulence genes (icaA, fbl, vwbl, fbpA, slush A, B and C, and genes of the putative ß-hemolysin and hemolysin III) and the ability to induce synergistic hemolytic activity and hemolysis after 24, 48 and 72 h were investigated in a collection of twenty-two S. lugdunensis clinical isolates. The collection of isolates, mainly from implant orthopedic infections, had previously been grouped by ribotyping/dendrogram analysis and studied for biofilm matrices, biomasses and antibiotic resistances. Two isolates, constituting a unique small ribogroup sharing the same cluster, exhibited an amplicon size of the slush operon (S. lugdunensis synergistic hemolysin) which was shorter than the expected 977 bp. This outcome can predict the genetic lineage of the S. lugdunensis strains. One isolate (cra1342) presented two deletions: one of 90 bp in slush A and the other of 91 bp in slush B. Another isolate (N860314) showed a single 193 bp deletion, which encompassed part of the slush B terminal sequence and most of slush C. The isolate N860314 was devoid of hemolytic activity after 24 h, and the first consideration was that the deleted region deals with the coding of the active enzymatic site of the slush hemolysin. On the other hand, cra1342 and N860314 isolates with different slush deletions and with hemolytic activity after 24 and 48 h, respectively, could have replaced the hemolytic phenotype through other processes.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus lugdunensis , Humanos , Staphylococcus lugdunensis/genética , Factores de Virulencia/genética , Proteínas Hemolisinas/genética , Hemólisis/genética , Operón , Infecciones Estafilocócicas/genética
5.
Nano Lett ; 21(18): 7595-7601, 2021 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-34469164

RESUMEN

Staphylococci bind to the blood protein von Willebrand Factor (vWF), thereby causing endovascular infections. Whether and how this interaction occurs with the medically important pathogen Staphylococcus epidermidis is unknown. Using single-molecule experiments, we demonstrate that the S. epidermidis protein Aap binds vWF via an ultrastrong force, ∼3 nN, the strongest noncovalent biological bond ever reported, and we show that this interaction is activated by tensile loading, suggesting a catch-bond behavior. Aap-vWF binding involves exclusively the A1 domain of vWF but requires both the A and B domains of Aap, as revealed by inhibition assays using specific monoclonal antibodies. Collectively, our results point to a mechanism where force-induced unfolding of the B repeats activates the A domain of Aap, shifting it from a weak- to a strong-binding state, which then engages into an ultrastrong interaction with vWF A1. This shear-dependent function of Aap offers promise for innovative antistaphylococcal therapies.


Asunto(s)
Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Staphylococcus epidermidis , Factor de von Willebrand , Fenómenos Mecánicos , Microscopía de Fuerza Atómica , Unión Proteica , Factor de von Willebrand/metabolismo
6.
Int J Mol Sci ; 23(11)2022 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-35682632

RESUMEN

The use of indwelling medical devices has constantly increased in recent years and has revolutionized the quality of life of patients affected by different diseases. However, despite the improvement of hygiene conditions in hospitals, implant-associated infections remain a common and serious complication in prosthetic surgery, mainly in the orthopedic field, where infection often leads to implant failure. Staphylococcus aureus is the most common cause of biomaterial-centered infection. Upon binding to the medical devices, these bacteria proliferate and develop dense communities encased in a protective matrix called biofilm. Biofilm formation has been proposed as occurring in several stages-(1) attachment; (2) proliferation; (3) dispersal-and involves a variety of host and staphylococcal proteinaceous and non-proteinaceous factors. Moreover, biofilm formation is strictly regulated by several control systems. Biofilms enable staphylococci to avoid antimicrobial activity and host immune response and are a source of persistent bacteremia as well as of localized tissue destruction. While considerable information is available on staphylococcal biofilm formation on medical implants and important results have been achieved on the treatment of biofilms, preclinical and clinical applications need to be further investigated. Thus, the purpose of this review is to gather current studies about the mechanism of infection of indwelling medical devices by S. aureus with a special focus on the biochemical factors involved in biofilm formation and regulation. We also provide a summary of the current therapeutic strategies to combat biomaterial-associated infections and highlight the need to further explore biofilm physiology and conduct research for innovative anti-biofilm approaches.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Materiales Biocompatibles/uso terapéutico , Biopelículas , Humanos , Calidad de Vida , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus/fisiología , Staphylococcus aureus/fisiología
7.
J Biol Chem ; 295(29): 10008-10022, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32499371

RESUMEN

Staphylococcus aureus is an important bacterial pathogen that can cause a wide spectrum of diseases in humans and other animals. S. aureus expresses a variety of virulence factors that promote infection with this pathogen. These include cell-surface proteins that mediate adherence of the bacterial cells to host extracellular matrix components, such as fibronectin and fibrinogen. Here, using immunoblotting, ELISA, and surface plasmon resonance analysis, we report that the iron-regulated surface determinant B (IsdB) protein, besides being involved in heme transport, plays a novel role as a receptor for the plasma and extracellular matrix protein vitronectin (Vn). Vn-binding activity was expressed by staphylococcal strains grown under iron starvation conditions when Isd proteins are expressed. Recombinant IsdB bound Vn dose dependently and specifically. Both near-iron transporter motifs NEAT1 and NEAT2 of IsdB individually bound Vn in a saturable manner, with KD values in the range of 16-18 nm Binding of Vn to IsdB was specifically blocked by heparin and reduced at high ionic strength. Furthermore, IsdB-expressing bacterial cells bound significantly higher amounts of Vn from human plasma than did an isdB mutant. Adherence to and invasion of epithelial and endothelial cells by IsdB-expressing S. aureus cells was promoted by Vn, and an αvß3 integrin-blocking mAb or cilengitide inhibited adherence and invasion by staphylococci, suggesting that Vn acts as a bridge between IsdB and host αvß3 integrin.


Asunto(s)
Proteínas de Transporte de Catión/química , Staphylococcus aureus/química , Vitronectina/química , Proteínas de Transporte de Catión/metabolismo , Humanos , Integrina alfaVbeta3/química , Integrina alfaVbeta3/metabolismo , Unión Proteica , Staphylococcus aureus/metabolismo , Vitronectina/metabolismo
8.
Nano Lett ; 20(12): 8919-8925, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33237786

RESUMEN

Binding of Staphylococcus aureus surface proteins to endothelial cell integrins plays essential roles in host cell adhesion and invasion, eventually leading to life-threatening diseases. The staphylococcal protein IsdB binds to ß3-containing integrins through a mechanism that has never been thoroughly investigated. Here, we identify and characterize at the nanoscale a previously undescribed stress-dependent adhesion between IsdB and integrin αVß3. The strength of single IsdB-αVß3 interactions is moderate (∼100 pN) under low stress, but it increases dramatically under high stress (∼1000-2000 pN) to exceed the forces traditionally reported for the binding between integrins and Arg-Gly-Asp (RGD) sequences. We suggest a mechanism where high mechanical stress induces conformational changes in the integrin from a low-affinity, weak binding state to a high-affinity, strong binding state. This single-molecule study highlights that direct adhesin-integrin interactions represent potential targets to fight staphylococcal infections.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Adhesinas Bacterianas/metabolismo , Proteínas de Transporte de Catión , Humanos , Proteínas de la Membrana/metabolismo , Unión Proteica
9.
J Biol Chem ; 294(10): 3588-3602, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30622139

RESUMEN

Staphylococcus aureus is a Gram-positive bacterium that can cause both superficial and deep-seated infections. Histones released by neutrophils kill bacteria by binding to the bacterial cell surface and causing membrane damage. We postulated that cell wall-anchored proteins protect S. aureus from the bactericidal effects of histones by binding to and sequestering histones away from the cell envelope. Here, we focused on S. aureus strain LAC and by using an array of biochemical assays, including surface plasmon resonance and ELISA, discovered that fibronectin-binding protein B (FnBPB) is the main histone receptor. FnBPB bound all types of histones, but histone H3 displayed the highest affinity and bactericidal activity and was therefore investigated further. H3 bound specifically to the A domain of recombinant FnBPB with a KD of 86 nm, ∼20-fold lower than that for fibrinogen. Binding apparently occurred by the same mechanism by which FnBPB binds to fibrinogen, because FnBPB variants defective in fibrinogen binding also did not bind H3. An FnBPB-deletion mutant of S. aureus LAC bound less H3 and was more susceptible to its bactericidal activity and to neutrophil extracellular traps, whereas an FnBPB-overexpressing mutant bound more H3 and was more resistant than the WT. FnBPB bound simultaneously to H3 and plasminogen, which after activation by tissue plasminogen activator cleaved the bound histone. We conclude that FnBPB provides a dual immune-evasion function that captures histones and prevents them from reaching the bacterial membrane and simultaneously binds plasminogen, thereby promoting its conversion to plasmin to destroy the bound histone.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Antiinfecciosos/farmacología , Histonas/farmacología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/metabolismo , Antiinfecciosos/metabolismo , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Histonas/metabolismo , Concentración Osmolar , Plasminógeno/metabolismo , Unión Proteica , Staphylococcus aureus/citología
10.
FASEB J ; 33(3): 4448-4457, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30566365

RESUMEN

Group B Streptococcus (GBS) colonizes the human lower intestinal and genital tracts and constitutes a major threat to neonates from pregnant carrier mothers and to adults with underlying morbidity. The pathogen expresses cell-surface virulence factors that enable cell adhesion and penetration and that counteract innate and adaptive immune responses. Among these, the complement interfering protein (CIP) was recently described for its capacity to interact with the human C4b ligand and to interfere with the classical- and lectin-complement pathways. In the present study, we provide evidence that CIP can also interact with C3, C3b, and C3d. Immunoassay-based competition experiments showed that binding of CIP to C3d interferes with the interaction between C3d and the complement receptor 2/cluster of differentiation 21 (CR2/CD21) receptor on B cells. By B-cell intracellular signaling assays, CIP was confirmed to down-regulate CR2/CD21-dependent B-cell activation. The CIP domain involved in C3d binding was mapped via hydrogen deuterium exchange-mass spectrometry. The data obtained reveal a new role for this GBS polypeptide at the interface between the innate and adaptive immune responses, adding a new member to the growing list of virulence factors secreted by gram-positive pathogens that incorporate multiple immunomodulatory functions.-Giussani, S., Pietrocola, G., Donnarumma, D., Norais, N., Speziale, P., Fabbrini, M., Margarit, I. The Streptococcus agalactiae complement interfering protein combines multiple complement-inhibitory mechanisms by interacting with both C4 and C3 ligands.


Asunto(s)
Proteínas Bacterianas/metabolismo , Complemento C3d/antagonistas & inhibidores , Complemento C4/antagonistas & inhibidores , Streptococcus agalactiae/patogenicidad , Factores de Virulencia/metabolismo , Secuencia de Aminoácidos , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Proteínas Bacterianas/farmacología , Sitios de Unión , Señalización del Calcio , Línea Celular Tumoral , Complemento C3b/antagonistas & inhibidores , Complemento C3b/metabolismo , Complemento C3d/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Activación de Linfocitos/efectos de los fármacos , Espectrometría de Masas , Unión Proteica , Mapeo de Interacción de Proteínas , Receptores de Complemento 3d/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Streptococcus agalactiae/inmunología , Streptococcus agalactiae/metabolismo , Resonancia por Plasmón de Superficie , Virulencia , Factores de Virulencia/farmacología
11.
Biofouling ; 36(1): 86-100, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31985269

RESUMEN

Staphylococcus lugdunensis is an emerging high-virulent pathogen causative of hospital-acquired infections. Biofilm formation is a complex pathogenic process that leads to well-established bacterial communities. There is a paucity of data on the composition of the biofilm matrix among S. lugdunensis strains. Here, twenty-two S. lugdunensis clinical isolates, mainly from orthopaedic infections but also from other clinical sources, were sub-grouped by ribotyping and dendrogram analysis. Biofilms were analysed by fluorimetric methods based on FITC-Wheat Germ Agglutinin, SYPRO Ruby and TOTO-1 dyes to detect exopolysaccharides, proteins and extracellular DNA (eDNA), respectively. Biofilm morphology was investigated under confocal laser scanning microscopy (CLSM). Isolates displayed intriguing diversities in biofilm mass and matrix composition. The content of exopolysaccharides was found to be to be strongly associated with the biofilm mass (R2 = 0.882), while the content of proteins turned out to be weakly (R2 = 0.465) and that of eDNA very weakly associated (R2 = 0.202) to the biofilm mass.


Asunto(s)
Proteínas Bacterianas/metabolismo , ADN Bacteriano/genética , Matriz Extracelular de Sustancias Poliméricas/metabolismo , Polisacáridos Bacterianos/metabolismo , Staphylococcus lugdunensis/crecimiento & desarrollo , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Farmacorresistencia Bacteriana/efectos de los fármacos , Farmacorresistencia Bacteriana/genética , Matriz Extracelular de Sustancias Poliméricas/genética , Humanos , Pruebas de Sensibilidad Microbiana , Microscopía Confocal , Polisacáridos Bacterianos/genética , Staphylococcus lugdunensis/efectos de los fármacos , Staphylococcus lugdunensis/metabolismo , Staphylococcus lugdunensis/ultraestructura
12.
Nano Lett ; 19(10): 7400-7410, 2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31532212

RESUMEN

Binding of the Staphylococcus aureus surface protein clumping factor A (ClfA) to endothelial cell integrin αVß3 plays a crucial role during sepsis, by causing endothelial cell apoptosis and loss of barrier integrity. ClfA uses the blood plasma protein fibrinogen (Fg) to bind to αVß3 but how this is achieved at the molecular level is not known. Here we investigate the mechanical strength of the three-component ClfA-Fg-αVß3 interaction on living bacteria, by means of single-molecule experiments. We find that the ClfA-Fg-αVß3 ternary complex is extremely stable, being able to sustain forces (∼800 pN) that are much stronger than those of classical bonds between integrins and the Arg-Gly-Asp (RGD) tripeptide sequence (∼100 pN). Adhesion forces between single bacteria and αVß3 are strongly inhibited by an anti-αVß3 antibody, the RGD peptide, and the cyclic RGD peptide cilengitide, showing that formation of the complex involves RGD-dependent binding sites and can be efficiently inhibited by αVß3 blockers. Collectively, our experiments favor a binding mechanism involving the extraordinary elasticity of Fg. In the absence of mechanical stress, RGD572-574 sequences in the Aα chains mediate weak binding to αVß3, whereas under high mechanical stress exposure of cryptic Aα chain RGD95-97 sequences leads to extremely strong binding to the integrin. Our results identify an unexpected and previously undescribed force-dependent binding mechanism between ClfA and αVß3 on endothelial cells, which could represent a potential target to fight staphylococcal bloodstream infections.


Asunto(s)
Coagulasa/metabolismo , Células Endoteliales/metabolismo , Fibrinógeno/metabolismo , Integrina alfaVbeta3/metabolismo , Staphylococcus aureus/fisiología , Adhesión Bacteriana , Células Endoteliales/microbiología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Unión Proteica , Estrés Mecánico
13.
Mol Microbiol ; 110(1): 82-94, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30030946

RESUMEN

Binding of microbial pathogens to host vitronectin (Vtn) is a common theme in the pathogenesis of invasive infections. In this study, we characterized the role of Vtn in the invasion of mucosal epithelial cells by Streptococcus agalactiae (i.e. group B streptococcus or GBS), a frequent human pathogen. Moreover, we identified PbsP, a previously described plasminogen-binding protein of GBS, as a dual adhesin that can also interact with human Vtn through its streptococcal surface repeat (SSURE) domains. Deletion of the pbsP gene decreases both bacterial adhesion to Vtn-coated inert surfaces and the ability of GBS to interact with epithelial cells. Bacterial adherence to and invasion of epithelial cells were either inhibited or enhanced by cell pretreatment with, respectively, anti-Vtn antibodies or Vtn, confirming the role of Vtn as a GBS ligand on host cells. Finally, antibodies directed against the integrin αv subunit inhibited Vtn-dependent cell invasion by GBS. Collectively, these results indicate that Vtn acts as a bridge between the SSURE domains of PbsP on the GBS surface and host integrins to promote bacterial invasion of epithelial cells. Therefore, inhibition of interactions between PbsP and extracellular matrix components could represent a viable strategy to prevent colonization and invasive disease by GBS.


Asunto(s)
Proteínas Bacterianas/metabolismo , Integrina alfaV/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/metabolismo , Streptococcus agalactiae/patogenicidad , Vitronectina/metabolismo , Células A549 , Adhesión Bacteriana/genética , Proteínas Bacterianas/genética , Células CACO-2 , Pared Celular/metabolismo , Células Epiteliales/microbiología , Humanos , Integrina alfaV/genética , Dominios Proteicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Streptococcus agalactiae/genética , Vitronectina/genética
14.
Proc Natl Acad Sci U S A ; 113(2): 410-5, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26715750

RESUMEN

Staphylococcus aureus surface protein SasG promotes cell-cell adhesion during the accumulation phase of biofilm formation, but the molecular basis of this interaction remains poorly understood. Here, we unravel the mechanical properties of SasG on the surface of living bacteria, that is, in its native cellular environment. Nanoscale multiparametric imaging of living bacteria reveals that Zn(2+) strongly increases cell wall rigidity and activates the adhesive function of SasG. Single-cell force measurements show that SasG mediates cell-cell adhesion via specific Zn(2+)-dependent homophilic bonds between ß-sheet-rich G5-E domains on neighboring cells. The force required to unfold individual domains is remarkably strong, up to ∼500 pN, thus explaining how SasG can withstand physiological shear forces. We also observe that SasG forms homophilic bonds with the structurally related accumulation-associated protein of Staphylococcus epidermidis, suggesting the possibility of multispecies biofilms during host colonization and infection. Collectively, our findings support a model in which zinc plays a dual role in activating cell-cell adhesion: adsorption of zinc ions to the bacterial cell surface increases cell wall cohesion and favors the projection of elongated SasG proteins away from the cell surface, thereby enabling zinc-dependent homophilic bonds between opposing cells. This work demonstrates an unexpected relationship between mechanics and adhesion in a staphylococcal surface protein, which may represent a general mechanism among bacterial pathogens for activating cell association.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Staphylococcus aureus/fisiología , Zinc/farmacología , Adhesión Bacteriana/efectos de los fármacos , Proteínas Bacterianas/química , Fenómenos Biomecánicos/efectos de los fármacos , Fenómenos Biofísicos/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Proteínas de la Membrana/química , Microscopía de Fuerza Atómica , Modelos Biológicos , Estructura Terciaria de Proteína , Staphylococcus aureus/efectos de los fármacos
15.
J Immunol ; 196(1): 385-94, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26608922

RESUMEN

The group B Streptococcus (GBS) is a leading cause of neonatal invasive disease. GBS bacteria are surrounded by a thick capsular polysaccharide that is a potent inhibitor of complement deposition via the alternative pathway. Several of its surface molecules can however activate the classical and lectin complement pathways, rendering this species still vulnerable to phagocytic killing. In this study we have identified a novel secreted protein named complement interfering protein (CIP) that downregulates complement activation via the classical and lectin pathways, but not the alternative pathway. The CIP protein showed high affinity toward C4b and inhibited its interaction with C2, presumably preventing the formation of the C4bC2a convertase. Addition of recombinant CIP to GBS cip-negative bacteria resulted in decreased deposition of C3b on their surface and in diminished phagocytic killing in a whole-blood assay. Our data reveal a novel strategy exploited by GBS to counteract innate immunity and could be valuable for the development of anti-infective agents against this important pathogen.


Asunto(s)
Proteínas Bacterianas/inmunología , Complemento C4b/inmunología , Vía Clásica del Complemento/inmunología , Lectina de Unión a Manosa de la Vía del Complemento/inmunología , Evasión Inmune/inmunología , Streptococcus agalactiae/inmunología , Adulto , Secuencia de Aminoácidos , Activación de Complemento/inmunología , Complemento C3b/biosíntesis , Complemento C3b/inmunología , Vía Alternativa del Complemento/inmunología , Vía Clásica del Complemento/efectos de los fármacos , Lectina de Unión a Manosa de la Vía del Complemento/efectos de los fármacos , Humanos , Inmunidad Innata , Datos de Secuencia Molecular , Fagocitosis/inmunología , Unión Proteica/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Streptococcus agalactiae/genética , Streptococcus agalactiae/metabolismo
16.
J Biol Chem ; 291(35): 18148-62, 2016 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-27387503

RESUMEN

Staphylococcus aureus is a commensal bacterium that has the ability to cause superficial and deep-seated infections. Like several other invasive pathogens, S. aureus can capture plasminogen from the human host where it can be converted to plasmin by host plasminogen activators or by endogenously expressed staphylokinase. This study demonstrates that sortase-anchored cell wall-associated proteins are responsible for capturing the bulk of bound plasminogen. Two cell wall-associated proteins, the fibrinogen- and fibronectin-binding proteins A and B, were found to bind plasminogen, and one of them, FnBPB, was studied in detail. Plasminogen captured on the surface of S. aureus- or Lactococcus lactis-expressing FnBPB could be activated to the potent serine protease plasmin by staphylokinase and tissue plasminogen activator. Plasminogen bound to recombinant FnBPB with a KD of 0.532 µm as determined by surface plasmon resonance. Plasminogen binding did not to occur by the same mechanism through which FnBPB binds to fibrinogen. Indeed, FnBPB could bind both ligands simultaneously indicating that their binding sites do not overlap. The N3 subdomain of FnBPB contains the full plasminogen-binding site, and this includes, at least in part, two conserved patches of surface-located lysine residues that were recognized by kringle 4 of the host protein.


Asunto(s)
Adhesinas Bacterianas/química , Proteínas Bacterianas/química , Plasminógeno/química , Staphylococcus aureus/química , Adhesinas Bacterianas/metabolismo , Proteínas Bacterianas/metabolismo , Humanos , Plasminógeno/metabolismo , Unión Proteica , Dominios Proteicos , Staphylococcus aureus/metabolismo
17.
Mol Microbiol ; 101(1): 27-41, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26888569

RESUMEN

Streptococcus agalactiae (Group B Streptococcus or GBS) is a leading cause of invasive infections in neonates whose virulence is dependent on its ability to interact with cells and host components. We here characterized a surface protein with a critical function in GBS pathophysiology. This adhesin, designated PbsP, possesses two Streptococcal Surface Repeat domains, a methionine and lysine-rich region, and a LPXTG cell wall-anchoring motif. PbsP mediates plasminogen (Plg) binding both in vitro and in vivo and we showed that cell surface-bound Plg can be activated into plasmin by tissue plasminogen activator to increase the bacterial extracellular proteolytic activity. Absence of PbsP results in a decreased bacterial transmigration across brain endothelial cells and impaired virulence in a murine model of infection. PbsP is conserved among the main GBS lineages and is a major plasminogen adhesin in non-CC17 GBS strains. Importantly, immunization of mice with recombinant PbsP confers protective immunity. Our results indicate that GBS have evolved different strategies to recruit Plg which indicates that the ability to acquire cell surface proteolytic activity is essential for the invasiveness of this bacterium.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Plasminógeno/metabolismo , Streptococcus agalactiae/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Bacteriana/fisiología , Pared Celular/metabolismo , Células Endoteliales/metabolismo , Fibrinolisina/metabolismo , Humanos , Ratones , Unión Proteica , Infecciones Estreptocócicas/microbiología , Streptococcus/metabolismo , Streptococcus agalactiae/genética , Streptococcus agalactiae/patogenicidad , Virulencia
18.
J Microencapsul ; 33(8): 750-762, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27845595

RESUMEN

The aim of this work was the assessment of the "in vivo" immune response of a poly(lactide-co-glycolide)-based nanoparticulate adjuvant for a sub-unit vaccine, namely, a purified recombinant collagen-binding bacterial adhesion fragment (CNA19), against Staphylococcus aureus-mediated infections. "In vivo" immunogenicity studies were performed on mice: immunisation protocols encompassed subcutaneous and intranasal administration of CNA19 formulated as nanoparticles (NPs) and furthermore, CNA19-loaded NPs formulated in a set-up thermosetting chitosan-ß-glycerolphosphate (chitosan-ß-GP) solution for intranasal route in order to extend antigen exposure to nasal mucosa. CNA19 loaded NPs (mean size of about 195 nm, 9.04 ± 0.37µg/mg as CNA19 loading capacity) confirmed as suitable vaccine for subcutaneous administration with a more pronounced adjuvant effect (about 3-fold higher) with respect to aluminium, recognised as "reference" adjuvant. CNA19 loaded NPs formulated in an optimised thermogelling chitosan-ß-GP solution showed promising results for eliciting an effective humoral response and a good chance as intranasal boosting dose.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Portadores de Fármacos/química , Ácido Láctico/química , Nanopartículas/química , Ácido Poliglicólico/química , Infecciones Estafilocócicas/prevención & control , Vacunas Estafilocócicas/administración & dosificación , Staphylococcus aureus/inmunología , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/uso terapéutico , Administración Intranasal , Animales , Femenino , Inmunidad Humoral , Ratones , Ratones Endogámicos BALB C , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Infecciones Estafilocócicas/inmunología , Vacunas Estafilocócicas/farmacología , Vacunas Estafilocócicas/uso terapéutico , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/farmacología , Vacunas Sintéticas/uso terapéutico
19.
J Biol Chem ; 289(30): 21003-21015, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24904056

RESUMEN

Streptococcus agalactiae (group B Streptococcus or GBS) is a common cause of invasive infections in newborn infants and adults. The ability of GBS to bind human fibrinogen is of crucial importance in promoting colonization and invasion of host barriers. We characterized here a novel fibrinogen-binding protein of GBS, designated FbsC (Gbs0791), which is encoded by the prototype GBS strain NEM316. FbsC, which bears two bacterial immunoglobulin-like tandem repeat domains and a C-terminal cell wall-anchoring motif (LPXTG), was found to be covalently linked to the cell wall by the housekeeping sortase A. Studies using recombinant FbsC indicated that it binds fibrinogen in a dose-dependent and saturable manner, and with moderate affinity. Expression of FbsC was detected in all clinical GBS isolates, except those belonging to the hypervirulent lineage ST17. Deletion of fbsC decreases NEM316 abilities to adhere to and invade human epithelial and endothelial cells, and to form biofilm in vitro. Notably, bacterial adhesion to fibrinogen and fibrinogen binding to bacterial cells were abolished following fbsC deletion in NEM316. Moreover, the virulence of the fbsC deletion mutant and its ability to colonize the brain were impaired in murine models of infection. Finally, immunization with recombinant FbsC significantly protected mice from lethal GBS challenge. In conclusion, FbsC is a novel fibrinogen-binding protein expressed by most GBS isolates that functions as a virulence factor by promoting invasion of epithelial and endothelial barriers. In addition, the protein has significant immunoprotective activity and may be a useful component of an anti-GBS vaccine.


Asunto(s)
Proteínas Bacterianas/inmunología , Fibrinógeno/inmunología , Interacciones Huésped-Patógeno/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae/fisiología , Factores de Virulencia/inmunología , Animales , Adhesión Bacteriana/genética , Adhesión Bacteriana/inmunología , Proteínas Bacterianas/genética , Células CACO-2 , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/microbiología , Células Endoteliales/patología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Células Epiteliales/patología , Fibrinógeno/genética , Humanos , Ratones , Unión Proteica/genética , Unión Proteica/inmunología , Infecciones Estreptocócicas/genética , Vacunas Estreptocócicas/genética , Vacunas Estreptocócicas/inmunología , Factores de Virulencia/genética
20.
Infect Immun ; 83(10): 4093-102, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26238710

RESUMEN

In this study, we investigated the cell wall-anchored fibronectin-binding proteins SpsD and SpsL from the canine commensal and pathogen Staphylococcus pseudintermedius for their role in promoting bacterial invasion of canine progenitor epidermal keratinocytes (CPEK). Invasion was examined by the gentamicin protection assay and fluorescence microscopy. An ΔspsD ΔspsL mutant of strain ED99 had a dramatically reduced capacity to invade CPEK monolayers, while no difference in the invasion level was observed with single mutants. Lactococcus lactis transformed with plasmids expressing SpsD and SpsL promoted invasion, showing that both proteins are important. Soluble fibronectin was required for invasion, and an RGD-containing peptide or antibodies recognizing the integrin α5ß1 markedly reduced invasion, suggesting an important role for the integrin in this process. Src kinase inhibitors effectively blocked internalization, suggesting a functional role for the kinase in invasion. In order to identify the minimal fibronectin-binding region of SpsD and SpsL involved in the internalization process, recombinant fragments of both proteins were produced. The SpsD520-846 and SpsL538-823 regions harboring the major fibronectin-binding sites inhibited S. pseudintermedius internalization. Finally, the effects of staphylococcal invasion on the integrity of different cell lines were examined. Because SpsD and SpsL are critical factors for adhesion and invasion, blocking these processes could provide a strategy for future approaches to treating infections.


Asunto(s)
Proteínas Bacterianas/metabolismo , Enfermedades de los Perros/microbiología , Células Epiteliales/microbiología , Fibronectinas/metabolismo , Infecciones Estafilocócicas/veterinaria , Staphylococcus/metabolismo , Animales , Proteínas Bacterianas/genética , Línea Celular , Pared Celular/genética , Pared Celular/metabolismo , Enfermedades de los Perros/metabolismo , Perros , Unión Proteica , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus/genética , Staphylococcus/patogenicidad , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA