Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Microb Drug Resist ; 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38770776

RESUMEN

Candidiasis is a common fungal infection caused by Candida species, with Candida albicans being the most prevalent. Resistance to azole drugs, commonly used to treat Candida infections, poses a significant challenge. Transcriptional activator candidate 1 (TAC1) gene has emerged as a key player in regulating drug resistance in C. albicans. This review explores the structure and function of the TAC1 gene and its role in azole resistance. This gene encodes a transcription factor that controls the expression of genes involved in drug resistance, such as efflux pump genes (CDR1, CDR2, and MDR1) and ERG11. Mutations in TAC1 can increase these genes' expression and confer resistance to azoles. Various TAC1 gene mutations, mostly gain-of-function mutations, have been identified, which upregulate CDR1 and CDR2 expression, resulting in azole resistance. Understanding the mechanisms of azole resistance mediated by the TAC1 gene is crucial for the strategies in the effective antifungal development pipeline.

2.
EMBO J ; 41(5): e109386, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35112724

RESUMEN

The mechanisms whereby neutrophils respond differentially to live and dead organisms are unknown. We show here that neutrophils produce 5- to 30-fold higher levels of the Cxcl2 chemokine in response to live bacteria, compared with killed bacteria or isolated bacterial components, despite producing similar levels of Cxcl1 or pro-inflammatory cytokines. Secretion of high levels of Cxcl2, which potently activates neutrophils by an autocrine mechanism, requires three signals. The first two signals are provided by two different sets of signal peptides released by live bacteria, which selectively activate formylated peptide receptor 1 (Fpr1) and Fpr2, respectively. Signal 3 originates from Toll-like receptor activation by microbial components present in both live and killed bacteria. Mechanistically, these signaling pathways converge at the level of the p38 MAP kinase, leading to activation of the AP-1 transcription factor and to Cxcl2 induction. Collectively, our data demonstrate that the simultaneous presence of agonists for Fpr1, Fpr2, and Toll-like receptors represents a unique signature associated with viable bacteria, which is sensed by neutrophils and induces Cxcl2-dependent autocrine cell activation.


Asunto(s)
Bacterias/metabolismo , Neutrófilos/metabolismo , Proteínas Proto-Oncogénicas c-fes/metabolismo , Receptores Toll-Like/metabolismo , Animales , Citocinas/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/fisiología
3.
Front Cell Infect Microbiol ; 11: 679792, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34568085

RESUMEN

Binding to plasminogen (Plg) enables bacteria to associate with and invade host tissues. The cell wall protein PbsP significantly contributes to the ability of group B streptococci, a frequent cause of invasive infection, to bind Plg. Here we sought to identify the molecular regions involved in the interactions between Plg and PbsP. The K4 Kringle domain of the Plg molecule was required for binding of Plg to whole PbsP and to a PbsP fragment encompassing a region rich in methionine and lysine (MK-rich domain). These interactions were inhibited by free L-lysine, indicating the involvement of lysine binding sites in the Plg molecule. However, mutation to alanine of all lysine residues in the MK-rich domain did not decrease its ability to bind Plg. Collectively, our data identify a novel bacterial sequence that can interact with lysine binding sites in the Plg molecule. Notably, such binding did not require the presence of lysine or other positively charged amino acids in the bacterial receptor. These data may be useful for developing alternative therapeutic strategies aimed at blocking interactions between group B streptococci and Plg.


Asunto(s)
Lisina , Plasminógeno , Sitios de Unión , Pared Celular/metabolismo , Lisina/metabolismo , Plasminógeno/metabolismo , Unión Proteica , Streptococcus agalactiae
4.
J Immunol ; 207(5): 1448-1455, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34362834

RESUMEN

Identification of the receptors involved in innate immune recognition of Staphylococcus aureus, a major cause of morbidity and mortality in humans, is essential to develop alternative strategies to treat infections caused by antibiotic-resistant strains. In the current study, we examine the role of endosomal TLRs, which sense the presence of prokaryotic-type nucleic acids, in anti-staphylococcal host defenses using infection models involving genetically defective mice. Single deficiencies in TLR7, 9, or 13 resulted in mild or no decrease in host defenses. However, the simultaneous absence of TLR7, 9, and 13 resulted in markedly increased susceptibility to cutaneous and systemic S. aureus infection concomitantly with decreased production of proinflammatory chemokines and cytokines, neutrophil recruitment to infection sites, and reduced production of reactive oxygen species. This phenotype was significantly more severe than that of mice lacking TLR2, which senses the presence of staphylococcal lipoproteins. Notably, the combined absence of TLR7, 9, and 13 resulted in complete abrogation of IL-12 p70 and IFN-ß responses to staphylococcal stimulation in macrophages. Taken together, our data highlight the presence of a highly integrated endosomal detection system, whereby TLR7, 9, and 13 cooperate in sensing the presence of staphylococcal nucleic acids. We demonstrate that the combined absence of these receptors cannot be compensated for by cell surface-associated TLRs, such as TLR2, or cytosolic receptors. These data may be useful to devise strategies aimed at stimulating innate immune receptors to treat S. aureus infections.


Asunto(s)
Endosomas/metabolismo , Glicoproteínas de Membrana/metabolismo , Neutrófilos/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/fisiología , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 9/metabolismo , Receptores Toll-Like/metabolismo , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Humanos , Inmunidad Innata , Mediadores de Inflamación/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Toll-Like 7/genética , Receptor Toll-Like 9/genética , Receptores Toll-Like/genética
5.
PLoS One ; 16(6): e0253242, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34129624

RESUMEN

Streptococcus agalactiae (group B streptococcus or GBS) is a commensal bacterium that can frequently behave as a pathogen, particularly in the neonatal period and in the elderly. The gut is a primary site of GBS colonization and a potential port of entry during neonatal infections caused by hypervirulent clonal complex 17 (CC17) strains. Here we studied the interactions between the prototypical CC17 BM110 strain and polarized enterocytes using the Caco-2 cell line. GBS could adhere to and invade these cells through their apical or basolateral surfaces. Basolateral invasion was considerably more efficient than apical invasion and predominated under conditions resulting in weakening of cell-to-cell junctions. Bacterial internalization occurred by a mechanism involving caveolae- and lipid raft-dependent endocytosis and actin re-organization, but not clathrin-dependent endocytosis. In the first steps of Caco-2 invasion, GBS colocalized with the early endocytic marker EEA-1, to later reside in acidic vacuoles. Taken together, these data suggest that CC17 GBS selectively adheres to the lateral surface of enterocytes from which it enters through caveolar lipid rafts using a classical, actin-dependent endocytic pathway. These data may be useful to develop alternative preventive strategies aimed at blocking GBS invasion of the intestinal barrier.


Asunto(s)
Enterocitos/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/patogenicidad , Adhesión Bacteriana , Células CACO-2/microbiología , Endocitosis , Humanos , Uniones Intercelulares/microbiología , Microscopía Fluorescente , Streptococcus agalactiae/fisiología , Virulencia
6.
Med Mycol ; 58(8): 1138-1148, 2020 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-32246714

RESUMEN

Members of the C. neoformans/C. gattiii species complex are an important cause of serious humans infections, including meningoencephalitis. We describe here a 45 kDa extracellular cellulase purified from culture supernatants of C. neoformans var. neoformans. The N-terminal sequence obtained from the purified protein was used to isolate a clone containing the full-length coding sequence from a C. neoformans var. neoformans (strain B-3501A) cDNA library. Bioinformatics analysis indicated that this gene is present, with variable homology, in all sequenced genomes of the C. neoformans/C. gattii species complex. The cDNA clone was used to produce a recombinant 45 kDa protein in E. coli that displayed the ability to convert carboxymethyl cellulose and was therefore designated as NG-Case (standing for Neoformans Gattii Cellulase). To explore its potential use as a vaccine candidate, the recombinant protein was used to immunize mice and was found capable of inducing T helper type 1 responses and delayed-type hypersensitivity reactions, but not immune protection against a highly virulent C. neoformans var grubii strain. These data may be useful to better understand the mechanisms underlying the ability C. neoformans/C. gattii to colonize plant habitats and to interact with the human host during infection.


Asunto(s)
Celulasa/inmunología , Cryptococcus/enzimología , Proteínas Fúngicas/inmunología , Animales , Carboximetilcelulosa de Sodio/metabolismo , Celulasa/química , Celulasa/genética , Celulasa/metabolismo , Criptococosis/inmunología , Criptococosis/microbiología , Cryptococcus/genética , Cryptococcus/inmunología , Cryptococcus/metabolismo , Cryptococcus neoformans/enzimología , Cryptococcus neoformans/genética , Cryptococcus neoformans/inmunología , Cryptococcus neoformans/metabolismo , Medios de Cultivo Condicionados , Citocinas/inmunología , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Inmunización , Ratones , Peso Molecular , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Células TH1/inmunología
7.
mBio ; 11(2)2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32209688

RESUMEN

Streptococcus pneumoniae (or pneumococcus) is a highly prevalent human pathogen. Toll-like receptors (TLRs) function as immune sensors that can trigger host defenses against this bacterium. Defects in TLR-activated signaling pathways, including deficiency in the adaptor protein myeloid differentiation factor 88 (MyD88), are associated with markedly increased susceptibility to infection. However, the individual MyD88-dependent TLRs predominantly involved in antipneumococcal defenses have not been identified yet. Here we find that triple knockout mice simultaneously lacking TLR7, TLR9, and TLR13, which sense the presence of bacterial DNA (TLR9) and RNA (TLR7 and TLR13) in the phagolysosomes of phagocytic cells, display a phenotype that largely resembles that of MyD88-deficient mice and rapidly succumb to pneumococcal pneumonitis due to defective neutrophil influx into the lung. Accordingly, TLR7/9/13 triple knockout resident alveolar macrophages were largely unable to respond to pneumococci with the production of neutrophil-attracting chemokines and cytokines. Mice with single deficiencies of TLR7, TLR9, or TLR13 showed unaltered ability to control lung infection but were moderately more susceptible to encephalitis, in association with a decreased ability of microglia to mount cytokine responses in vitro Our data point to a dominant, tissue-specific role of nucleic acid-sensing pathways in innate immune recognition of S. pneumoniae and also show that endosomal TLRs are largely capable of compensating for the absence of each other, which seems crucial to prevent pneumococci from escaping immune recognition. These results may be useful to develop novel strategies to treat infections by antibiotic-resistant pneumococci based on stimulation of the innate immune system.IMPORTANCE The pneumococcus is a bacterium that frequently causes infections in the lungs, ears, sinus cavities, and meninges. During these infections, body defenses are triggered by tissue-resident cells that use specialized receptors, such as Toll-like receptors (TLRs), to sense the presence of bacteria. We show here that pneumococci are predominantly detected by TLRs that are located inside intracellular vacuoles, including endosomes, where these receptors can sense the presence of nucleic acids released from ingested bacteria. Mice that simultaneously lacked three of these receptors (specifically, TLR7, TLR9, and TLR13) were extremely susceptible to lung infection and rapidly died after inhalation of pneumococci. Moreover, tissue-resident macrophages from these mice were impaired in their ability to respond to the presence of pneumococci by producing inflammatory mediators capable of recruiting polymorphonuclear leucocytes to infection sites. This information may be useful to develop drugs to treat pneumococcal infections, particularly those caused by antibiotic-resistant strains.


Asunto(s)
Inmunidad Innata , Ácidos Nucleicos/inmunología , Streptococcus pneumoniae/inmunología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Animales , Citocinas/inmunología , Femenino , Pulmón/inmunología , Pulmón/microbiología , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Infiltración Neutrófila , Transducción de Señal , Streptococcus pneumoniae/patogenicidad , Receptor Toll-Like 7/genética , Receptor Toll-Like 9/genética
8.
J Immunol ; 204(3): 660-670, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31852751

RESUMEN

The influx of neutrophils to infection sites is a fundamental step in host defenses against the frequent human pathogen group B Streptococcus (GBS) and other extracellular bacteria. Using a mouse model of GBS-induced peritonitis, we show in this study that the chemokines Cxcl1 and Cxcl2 play distinctive roles in enhancing the recruitment and the antibacterial activities of neutrophils in a manner that is linked to differences in the cellular sources of these mediators. Cell depletion experiments demonstrated that neutrophils make a significant contribution to the in vivo production of Cxcl2 but not Cxcl1. In vitro, neutrophils responded weakly to LPS but released high levels of Cxcl2 after stimulation with GBS or other bacteria. Neutrophil-derived Cxcl2 acted in an autocrinous manner to increase its own production and to enhance antibacterial activities, including the release of oxygen radicals. In both neutrophils and macrophages, the production of Cxcl1/2 largely required the presence of functional UNC93B1, a chaperone protein involved in signaling by endosomal TLRs. Moreover, the phenotype of UNC93B1-defective phagocytes could be recapitulated by the simultaneous absence of TLR7, 9, and 13 but not by the absence of individual TLRs. Collectively, our data show that neutrophils recognize Gram-positive and Gram-negative bacteria by means of multiple phagosomal TLRs, resulting in de novo synthesis of Cxcl2, amplification of neutrophil recruitment, and potentiation of their antibacterial activities. These data may be useful to devise alternative therapeutic strategies aimed at enhancing the recruitment and the functional activities of polymorphonuclear leukocytes during infections caused by antibiotic-resistant bacteria.


Asunto(s)
Infecciones Bacterianas/inmunología , Quimiocina CXCL2/metabolismo , Endosomas/metabolismo , Neutrófilos/inmunología , Peritonitis/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Receptores Toll-Like/metabolismo
9.
Sci Rep ; 8(1): 14322, 2018 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-30254272

RESUMEN

Streptococcus agalactiae (Group B Streptococcus or GBS) is a frequent cause of serious disease in newborns and adults. Epidemiological evidence indicates a strong association between GBS strains belonging to the hypervirulent CC17 clonal complex and the occurrence of meningitis in neonates. We investigate here the role of PbsP, a cell wall plasminogen binding protein, in colonization of the central nervous system by CC17 GBS. Deletion of pbsP selectively impaired the ability of the CC17 strain BM110 to colonize the mouse brain after intravenous challenge, despite its unchanged capacity to persist at high levels in the blood and to invade the kidneys. Moreover, immunization with a recombinant form of PbsP considerably reduced brain infection and lethality. In vitro, pbsP deletion markedly decreased plasmin-dependent transmigration of BM110 through brain microvascular endothelial cells. Although PbsP was modestly expressed in bacteria grown under standard laboratory conditions, pbsP expression was markedly upregulated during in vivo infection or upon contact with cultured brain endothelial cells. Collectively, our studies indicate that PbsP is a highly conserved Plg binding adhesin, which is functionally important for invasion of the central nervous system by the hypervirulent CC17 GBS. Moreover, this antigen is a promising candidate for inclusion in a universal GBS vaccine.


Asunto(s)
Proteínas Bacterianas/metabolismo , Encéfalo/microbiología , Streptococcus agalactiae/metabolismo , Streptococcus agalactiae/patogenicidad , Animales , Encéfalo/citología , Movimiento Celular , Células Endoteliales/citología , Fibrinolisina/metabolismo , Regulación Bacteriana de la Expresión Génica , Ratones , Streptococcus agalactiae/genética , Virulencia
10.
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
11.
PLoS One ; 11(8): e0160702, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27508302

RESUMEN

We explore here the potential of a newly described technology, which is named PROFILER and is based on next generation sequencing of gene-specific lambda phage-displayed libraries, to rapidly and accurately map monoclonal antibody (mAb) epitopes. For this purpose, we used a novel mAb (designated 31E10/E7) directed against Neisserial Heparin-Binding Antigen (NHBA), a component of the anti-group B meningococcus Bexsero® vaccine. An NHBA phage-displayed library was affinity-selected with mAb 31E10/E7, followed by massive sequencing of the inserts present in antibody-selected phage pools. Insert analysis identified an amino acid stretch (D91-A128) in the N-terminal domain, which was shared by all of the mAb-enriched fragments. Moreover, a recombinant fragment encompassing this sequence could recapitulate the immunoreactivity of the entire NHBA molecule against mAb 31E10/E7. These results were confirmed using a panel of overlapping recombinant fragments derived from the NHBA vaccine variant and a set of chemically synthetized peptides covering the 10 most frequent antigenic variants. Furthermore, hydrogen-deuterium exchange mass-spectrometry analysis of the NHBA-mAb 31E10/E7 complex was also compatible with mapping of the epitope to the D91-A128 region. Collectively, these results indicate that the PROFILER technology can reliably identify epitope-containing antigenic fragments and requires considerably less work, time and reagents than other epitope mapping methods.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Portadoras/inmunología , Mapeo Epitopo/métodos , Biblioteca de Péptidos , Animales , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas Portadoras/genética , Reacciones Cruzadas , Secuenciación de Nucleótidos de Alto Rendimiento , Espectrometría de Masas/métodos , Ratones , Neisseria meningitidis Serogrupo B/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología
12.
PLoS One ; 11(8): e0160249, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27509078

RESUMEN

Previous studies have shown that the pro-inflammatory cytokine IL-1ß has a crucial role in host defenses against group B streptococcus (GBS), a frequent human pathogen, by recruiting neutrophils to infection sites. We examined here the cell types and mechanisms involved in IL-1ß production during infection. Using a GBS-induced peritonitis model in mice, we first found that a large proportion of exudate cells contain intracellular IL-1ß by immunofluorescence. Of the IL-1ß positive cells, 82 and 7% were neutrophils and macrophages, respectively, suggesting that the former cell type might significantly contribute to IL-1ß production. Accordingly, depletion of neutrophils with anti-Ly6G antibodies resulted in a significant reduction in the levels of IL-1ß, but not of TNF-α or IL-6. We next found that neutrophils are capable of releasing mature IL-1ß and TNF-α directly in response to in vitro stimulation with GBS. The production of pro-IL-1ß and TNF-α in these cells required the Toll-like receptor (TLR) adaptor MyD88 and the chaperone protein UNC93B1, which is involved in mobilization of a subfamily of TLRs to the endosomes. Moreover, pro-IL-1ß processing and IL-1ß release was triggered by GBS hemolysin and required components of the canonical inflammasome, including caspase-1, ASC and NLRP3. Collectively our findings indicate that neutrophils make a significant contribution to IL-1ß production during GBS infection, thereby amplifying their own recruitment. These cells directly recognize GBS by means of endosomal TLRs and cytosolic sensors, leading to activation of the caspase-1 inflammasome.


Asunto(s)
Interleucina-1beta/metabolismo , Neutrófilos/metabolismo , Streptococcus/fisiología , Animales , Anticuerpos/inmunología , Antígenos Ly/inmunología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Caspasa 1/deficiencia , Caspasa 1/genética , Caspasa 1/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteínas Hemolisinas/inmunología , Proteínas Hemolisinas/metabolismo , Interleucina-1beta/análisis , Interleucina-6/análisis , Interleucina-6/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neutrófilos/inmunología , Peritonitis/inmunología , Peritonitis/microbiología , Peritonitis/patología , Serogrupo , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/patología , Streptococcus/aislamiento & purificación , Receptores Toll-Like/metabolismo , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/metabolismo
13.
Sci Rep ; 6: 31458, 2016 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-27530334

RESUMEN

We have recently described a method, named PROFILER, for the identification of antigenic regions preferentially targeted by polyclonal antibody responses after vaccination. To test the ability of the technique to provide insights into the functional properties of monoclonal antibody (mAb) epitopes, we used here a well-characterized epitope of meningococcal factor H binding protein (fHbp), which is recognized by mAb 12C1. An fHbp library, engineered on a lambda phage vector enabling surface expression of polypeptides of widely different length, was subjected to massive parallel sequencing of the phage inserts after affinity selection with the 12C1 mAb. We detected dozens of unique antibody-selected sequences, the most enriched of which (designated as FrC) could largely recapitulate the ability of fHbp to bind mAb 12C1. Computational analysis of the cumulative enrichment of single amino acids in the antibody-selected fragments identified two overrepresented stretches of residues (H248-K254 and S140-G154), whose presence was subsequently found to be required for binding of FrC to mAb 12C1. Collectively, these results suggest that the PROFILER technology can rapidly and reliably identify, in the context of complex conformational epitopes, discrete "hot spots" with a crucial role in antigen-antibody interactions, thereby providing useful clues for the functional characterization of the epitope.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/química , Bacteriófago lambda/genética , Epítopos/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Biblioteca de Péptidos , Animales , Ratones
14.
MAbs ; 8(4): 741-50, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26963435

RESUMEN

There is a strong need for rapid and reliable epitope mapping methods that can keep pace with the isolation of increasingly larger numbers of mAbs. We describe here the identification of a conformational epitope using Phage-based Representation OF ImmunoLigand Epitope Repertoire (PROFILER), a recently developed high-throughput method based on deep sequencing of antigen-specific lambda phage-displayed libraries. A novel bactericidal monoclonal antibody (mAb 9F11) raised against Neisseria meningitidis adhesin A (NadA), an important component of the Bexsero(®) anti-meningococcal vaccine, was used to evaluate the technique in comparison with other epitope mapping methods. The PROFILER technology readily identified NadA fragments that were capable of fully recapitulating the reactivity of the entire antigen against mAb 9F11. Further analysis of these fragments using mutagenesis and hydrogen-deuterium exchange mass-spectrometry allowed us to identify the binding site of mAb 9F11 (A250-D274) and an adjoining sequence (V275-H312) that was also required for the full functional reconstitution of the epitope. These data suggest that, by virtue of its ability to detect a great variety of immunoreactive antigen fragments in phage-displayed libraries, the PROFILER technology can rapidly and reliably identify epitope-containing regions and provide, in addition, useful clues for the functional characterization of conformational mAb epitopes.


Asunto(s)
Adhesinas Bacterianas/inmunología , Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Técnicas de Visualización de Superficie Celular/métodos , Mapeo Epitopo/métodos , Neisseria meningitidis/inmunología , Animales , Vacunas Bacterianas/inmunología , Vacunas Meningococicas , Fragmentos de Péptidos/inmunología
15.
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
16.
J Proteomics ; 89: 154-64, 2013 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-23770297

RESUMEN

The two-component regulatory system CovRS is the main regulator of virulence gene expression in Group B Streptococcus (GBS), the leading cause of invasive infections in neonates. In this study we analyzed by mass spectrometry the GBS extracellular protein complex (i.e. the exoproteome) of NEM316 wild-type (WT) strain and its isogenic covRS deletion mutant (ΔcovRS). A total of 53 proteins, 49 of which had classical secretion signals, were identified: 12 were released by both strains while 21 and 20 were released exclusively by WT and ΔcovRS strains, respectively. In addition to known surface proteins, we detected here unstudied cell-wall associated proteins and/or orthologs of putative virulence factors present in other pathogenic streptococci. While the functional role of these proteins remains to be elucidated, our data suggest that the analysis of the exoproteome of bacterial pathogens under different gene expression conditions may be a powerful tool for the rapid identification of novel virulence factors and vaccine candidates. BIOLOGICAL SIGNIFICANCE: We believe that this manuscript will be of interest to Journal of Proteomics readers since the paper describes the identification of several putative virulence factors and vaccine candidates of the group B streptococcus, an important pathogen, using a simple proteomics strategy involving LC-MS analysis of culture supernatants obtained from two strains with divergent gene expression patterns. This technique provided the most comprehensive inventory of extracellular proteins obtained from a single streptococcal species thus far. The approach described has the added benefit of being easily applicable to a large number of different strains, making it ideal for the identification of conserved vaccine candidates.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteoma/metabolismo , Streptococcus agalactiae/metabolismo , Factores de Virulencia/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Espectrometría de Masas , Proteoma/inmunología , Vacunas Estreptocócicas/genética , Vacunas Estreptocócicas/inmunología , Streptococcus agalactiae/genética , Streptococcus agalactiae/inmunología , Factores de Virulencia/genética , Factores de Virulencia/inmunología
17.
Eur J Immunol ; 42(10): 2632-43, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22777843

RESUMEN

Despite convincing evidence for involvement of members of the Toll-like receptor (TLR) family in fungal recognition, little is known of the functional role of individual TLRs in antifungal defenses. We found here that TLR7 was partially required for the induction of IL-12 (IL-12p70) by Candida albicans or Saccharomyces cerevisiae. Moreover, the IL-12p70 response was completely abrogated in cells from 3d mice, which are unable to mobilize TLRs to endosomal compartments, as well as in cells from mice lacking either the TLR adaptor MyD88 or the IRF1 transcription factor. Notably, purified fungal RNA recapitulated IL-12p70 induction by whole yeast. Although RNA could also induce moderate TLR7-dependent IL-23 and tumor necrosis factor-alpha (TNF-α) secretion, TLR7 and other endosomal TLRs were redundant for IL-23 or TNF-α induction by whole fungi. Importantly, mice lacking TLR7 or IRF1 were hypersusceptible to systemic C. albicans infection. Our data suggest that IRF1 is downstream of a novel, nonredundant fungal recognition pathway that has RNA as a major target and requires phagosomal recruitment of intracellular TLRs. This pathway differs from those involved in IL-23 or TNF-α responses, which we show here to be independent from translocation of intracellular TLRs, phagocytosis, or phagosomal acidification.


Asunto(s)
Candida albicans/inmunología , Candidiasis/inmunología , Células Dendríticas/inmunología , ARN de Hongos/inmunología , Animales , Candida albicans/genética , Citocinas/metabolismo , Células Dendríticas/microbiología , Susceptibilidad a Enfermedades , Endosomas/genética , Endosomas/metabolismo , Inmunidad , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Fagocitosis/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/inmunología , Receptor Toll-Like 7/genética
18.
PLoS One ; 7(5): e36588, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22570729

RESUMEN

There is considerable interest in pneumococcal protein antigens capable of inducing serotype-independent immunoprotection and of improving, thereby, existing vaccines. We report here on the immunogenic properties of a novel surface antigen encoded by ORF spr1875 in the R6 strain genome. An antigenic fragment encoded by spr1875, designated R4, was identified using a Streptococcus pneumoniae phage displayed genomic library after selection with a human convalescent serum. Immunofluorescence analysis with anti-R4 antisera showed that Spr1875 was expressed on the surface of strains belonging to different serotypes. Moreover, the gene was present with little sequence variability in 27 different pneumococcal strains isolated worldwide. A mutant lacking Spr1875 was considerably less virulent than the wild type D39 strain in an intravenous mouse model of infection. Moreover, immunization with the R4 recombinant fragment, but not with the whole Spr1875 protein, induced significant protection against sepsis in mice. Lack of protection after immunization with the whole protein was related to the presence of immunodominant, non-protective epitopes located outside of the R4 fragment. In conclusion, our data indicate that Spr1875 has a role in pneumococcal virulence and is immunogenic. As the R4 fragment conferred immunoprotection from experimental sepsis, selected antigenic fragments of Spr1875 may be useful for the development of a pneumococcal protein-based vaccine.


Asunto(s)
Proteínas Bacterianas/inmunología , Biblioteca de Péptidos , Infecciones Neumocócicas/prevención & control , Streptococcus pneumoniae/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/química , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Mutación , Péptidos/inmunología , Infecciones Neumocócicas/mortalidad , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Serotipificación , Streptococcus pneumoniae/clasificación
19.
J Immunol ; 188(4): 1953-60, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22250086

RESUMEN

Group B Streptococcus (GBS) is a frequent agent of life-threatening sepsis and meningitis in neonates and adults with predisposing conditions. We tested the hypothesis that activation of the inflammasome, an inflammatory signaling complex, is involved in host defenses against this pathogen. We show in this study that murine bone marrow-derived conventional dendritic cells responded to GBS by secreting IL-1ß and IL-18. IL-1ß release required both pro-IL-1ß transcription and caspase-1-dependent proteolytic cleavage of intracellular pro-IL-1ß. Dendritic cells lacking the TLR adaptor MyD88, but not those lacking TLR2, were unable to produce pro-IL-1ß mRNA in response to GBS. Pro-IL-1ß cleavage and secretion of the mature IL-1ß form depended on the NOD-like receptor family, pyrin domain containing 3 (NLRP3) sensor and the apoptosis-associated speck-like protein containing a caspase activation and recruitment domain adaptor. Moreover, activation of the NLRP3 inflammasome required GBS expression of ß-hemolysin, an important virulence factor. We further found that mice lacking NLRP3, apoptosis-associated speck-like protein, or caspase-1 were considerably more susceptible to infection than wild-type mice. Our data link the production of a major virulence factor by GBS with the activation of a highly effective anti-GBS response triggered by the NLRP3 inflammasome.


Asunto(s)
Proteínas Portadoras/inmunología , Proteínas Portadoras/metabolismo , Inflamasomas/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae/inmunología , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Bacterianas/biosíntesis , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/genética , Caspasa 1/genética , Caspasa 1/metabolismo , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Femenino , Proteínas Hemolisinas/biosíntesis , Interleucina-18/biosíntesis , Interleucina-18/metabolismo , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Proteína con Dominio Pirina 3 de la Familia NLR , ARN Mensajero/biosíntesis , Transducción de Señal , Infecciones Estreptocócicas/metabolismo , Streptococcus agalactiae/metabolismo , Streptococcus agalactiae/patogenicidad , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética
20.
Eur J Immunol ; 41(7): 1969-79, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21480215

RESUMEN

Although type I interferons (IFN-α/ß) have been traditionally associated with antiviral responses, their importance in host defense against bacterial pathogens is being increasingly appreciated. Little is known, however, about the occurrence and functional role of IFN-α/ß production in response to pathogenic yeasts. Here, we found that conventional DCs, but not macrophages nor plasmacytoid DCs, mounted IFN-ß responses after in vitro stimulation with Candida spp. or Saccharomyces cerevisiae. These responses absolutely required MyD88, a Toll-like receptor (TLR) adaptor molecule, and were partially dependent on TLR9 and TLR7. Moreover, Candida DNA, as well as RNA, could recapitulate the IFN-ß response. After intravenous challenge with Candida albicans, most mice lacking the IFN-α/ß receptor died from their inability to control fungal growth, whereas all WT controls survived. These data suggest that recognition of yeast nucleic acids by TLR7 and TLR9 triggers a host-protective IFN-α/ß response.


Asunto(s)
Candida albicans/inmunología , Candidiasis/inmunología , ADN de Hongos/inmunología , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , ARN de Hongos/inmunología , Saccharomyces cerevisiae/inmunología , Animales , Candida albicans/genética , Células Dendríticas/inmunología , Células HEK293 , Humanos , Interferón Tipo I/genética , Interferón beta/inmunología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , Reacción en Cadena de la Polimerasa , Saccharomyces cerevisiae/genética , Transducción de Señal , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...