Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
BMC Microbiol ; 24(1): 221, 2024 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-38909237

RESUMEN

BACKGROUND: Group B Streptococcus (GBS) is a commensal of healthy adults and an important pathogen in newborns, the elderly and immunocompromised individuals. GBS displays several virulence factors that promote colonisation and host infection, including the ST-17 strain-specific adhesin Srr2, previously characterised for its binding to fibrinogen. Another common target for bacterial adhesins and for host colonization is fibronectin, a multi-domain glycoprotein found ubiquitously in body fluids, in the extracellular matrix and on the surface of cells. RESULTS: In this study, fibronectin was identified as a novel ligand for the Srr2 adhesin of GBS. A derivative of the ST-17 strain BM110 overexpressing the srr2 gene showed an increased ability to bind fibrinogen and fibronectin, compared to the isogenic wild-type strain. Conversely, the deletion of srr2 impaired bacterial adhesion to both ligands. ELISA assays and surface plasmon resonance studies using the recombinant binding region (BR) form of Srr2 confirmed a direct interaction with fibronectin with an estimated Kd of 92 nM. Srr2-BR variants defective in fibrinogen binding also exhibited no interaction with fibronectin, suggesting that Srr2 binds this ligand through the dock-lock-latch mechanism, previously described for fibrinogen binding. The fibronectin site responsible for recombinant Srr2-BR binding was identified and localised in the central cell-binding domain of the protein. Finally, in the presence of fibronectin, the ability of a Δsrr2 mutant to adhere to human cervico-vaginal epithelial cells was significantly lower than that of the wild-type strain. CONCLUSION: By combining genetic and biochemical approaches, we demonstrate a new role for Srr2, namely interacting with fibronectin. We characterised the molecular mechanism of this interaction and demonstrated that it plays a role in promoting the adhesion of GBS to human cervico-vaginal epithelial cells, further substantiating the role of Srr2 as a factor responsible for the hypervirulence of GBS ST-17 strains. The discovery of the previously undescribed interaction between Srr2 and fibronectin establishes this adhesin as a key factor for GBS colonisation of host tissues.


Asunto(s)
Adhesinas Bacterianas , Adhesión Bacteriana , Fibronectinas , Unión Proteica , Streptococcus agalactiae , Streptococcus agalactiae/genética , Streptococcus agalactiae/metabolismo , Streptococcus agalactiae/patogenicidad , Fibronectinas/metabolismo , Humanos , Adhesinas Bacterianas/metabolismo , Adhesinas Bacterianas/genética , Fibrinógeno/metabolismo , Fibrinógeno/genética , Células Epiteliales/microbiología , Femenino , Infecciones Estreptocócicas/microbiología , Factores de Virulencia/metabolismo , Factores de Virulencia/genética
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.
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.
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
6.
Nat Immunol ; 10(6): 587-94, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19430477

RESUMEN

Little is known of how and where bacterial recognition triggers the induction of type I interferon. Whether the type of recognition receptor used in these responses is determined by the subcellular location of bacteria is not understood. Here we show that phagosomal bacteria such as group B streptococcus, but not cytosolic bacteria, potently induced interferon in conventional dendritic cells by a mechanism that required Toll-like receptor 7, the adaptor MyD88 and the transcription factor IRF1, all of which localized together with bacterial products in degradative vacuoles bearing lysosomal markers. Thus, this cell type-specific recognition pathway links lysosomal recognition of bacterial RNA with a robust, host-protective interferon response.


Asunto(s)
Células Dendríticas/metabolismo , Lisosomas/metabolismo , Glicoproteínas de Membrana/metabolismo , Streptococcus agalactiae/inmunología , Receptor Toll-Like 7/metabolismo , Animales , Animales Recién Nacidos/inmunología , Animales Recién Nacidos/microbiología , Células Dendríticas/inmunología , Femenino , Factor 1 Regulador del Interferón/inmunología , Factor 1 Regulador del Interferón/metabolismo , Interferón beta/biosíntesis , Lisosomas/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Fagocitosis , Fagosomas/inmunología , Fagosomas/metabolismo , ARN Bacteriano/metabolismo , Transducción de Señal , Infecciones Estreptocócicas/inmunología , Receptor Toll-Like 7/inmunología
7.
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
8.
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
9.
Gut ; 67(3): 542-552, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28428345

RESUMEN

OBJECTIVE: The stability of the covalently closed circular DNA (cccDNA) in nuclei of non-dividing hepatocytes represents a key determinant of HBV persistence. Contrarily, studies with animal hepadnaviruses indicated that hepatocyte turnover can reduce cccDNA loads but knowledge on the proliferative capacity of HBV-infected primary human hepatocytes (PHHs) in vivo and the fate of cccDNA in dividing PHHs is still lacking. This study aimed to determine the impact of human hepatocyte division on cccDNA stability in vivo. METHODS: PHH proliferation was triggered by serially transplanting hepatocytes from HBV-infected humanised mice into naïve recipients. Cell proliferation and virological changes were assessed by quantitative PCR, immunofluorescence and RNA in situ hybridisation. Viral integrations were analysed by gel separation and deep sequencing. RESULTS: PHH proliferation strongly reduced all infection markers, including cccDNA (median 2.4 log/PHH). Remarkably, cell division appeared to cause cccDNA dilution among daughter cells and intrahepatic cccDNA loss. Nevertheless, HBV survived in sporadic non-proliferating human hepatocytes, so that virological markers rebounded as hepatocyte expansion relented. This was due to reinfection of quiescent PHHs since treatment with the entry inhibitor myrcludex-B or nucleoside analogues blocked viral spread and intrahepatic cccDNA accumulation. Viral integrations were detected both in donors and recipient mice but did not appear to contribute to antigen production. CONCLUSIONS: We demonstrate that human hepatocyte division even without involvement of cytolytic mechanisms triggers substantial cccDNA loss. This process may be fundamental to resolve self-limiting acute infection and should be considered in future therapeutic interventions along with entry inhibition strategies.


Asunto(s)
Proliferación Celular , ADN Circular/metabolismo , ADN Viral/metabolismo , Virus de la Hepatitis B/fisiología , Hepatitis B Crónica , Hepatocitos/fisiología , Animales , División Celular , Quimera , Modelos Animales de Enfermedad , Antígenos del Núcleo de la Hepatitis B/metabolismo , Antígenos de Superficie de la Hepatitis B/sangre , Virus de la Hepatitis B/genética , Hepatitis B Crónica/prevención & control , Humanos , Queratina-18/metabolismo , Lamivudine/uso terapéutico , Lipopéptidos/uso terapéutico , Ratones , Cultivo Primario de Células , Recurrencia , Inhibidores de la Transcriptasa Inversa/uso terapéutico , Carga Viral , Integración Viral , Replicación Viral
10.
Intervirology ; 61(1): 1-8, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30021203

RESUMEN

BACKGROUND: Hepatitis C virus (HCV) NS3 resistance-associated substitutions (RASs) reduce HCV susceptibility to protease inhibitors. Little is known about NS3 RASs in viral isolates from the liver of chronic hepatitis C (CHC) patients infected with HCV genotype-1a (G1a). AIM: The objective of this work was to study NS3 variability in isolates from the serum and liver of HCV-G1a-infected patients naïve to direct-acting antivirals (DAAs). METHODS: NS3 variability of HCV-G1a isolates from the serum and liver of 11 naïve CHC patients, and from sera of an additional 20 naïve CHC patients, was investigated by next-generation sequencing. RESULTS: At a cutoff of 1%, NS3 RASs were detected in all the samples examined. At a cutoff of 15%, they were found in 54.5% (6/11) and 27.3% (3/11) of the paired liver and serum samples, respectively, and in 22.5% (7/31) of the overall serum samples examined. Twenty-six out of thirty-one (84%) patients showed NS3 variants with multiple RASs. Phylogenetic analysis showed that NS3 sequences clustered within 2 clades, with 10/31 (32.2%) patients infected by clade I, 15/31 (48.8%) by clade II, and 6/31 (19.3%) by both clades. CONCLUSIONS: Though the number of patients examined was limited, NS3 variants with RASs appear to be major components of both intrahepatic and circulating viral quasispecies populations in DAA-naïve patients.


Asunto(s)
Variación Genética , Hepacivirus/enzimología , Hepatitis C Crónica/virología , Proteínas no Estructurales Virales/genética , Adulto , Sustitución de Aminoácidos , Antivirales/farmacología , Farmacorresistencia Viral , Femenino , Genotipo , Hepacivirus/genética , Hepatitis C Crónica/epidemiología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Italia/epidemiología , Hígado/virología , Masculino , Persona de Mediana Edad , Filogenia , Inhibidores de Proteasas/farmacología , Suero/virología
11.
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
12.
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
13.
J Immunol ; 191(4): 1873-82, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23863905

RESUMEN

Newborns and infants present a higher susceptibility to infection than adults, a vulnerability associated with deficiencies in both the innate and adaptive immune systems. Innate immune receptors are sensors involved in the recognition and elimination of microbes that play a pivotal role at the interface between innate and adaptive immunity. Pentraxin 3 (PTX3), the prototypic long pentraxin, is a soluble pattern recognition receptor involved in the initiation of protective responses against selected pathogens. Because neonates are generally resistant to these pathogens, we suspected that PTX3 may be provided by a maternal source during the early life times. We observed that human colostrum contains high levels of PTX3, and that mammary epithelial cell and CD11b(+) milk cells constitutively produce PTX3. Interestingly, PTX3 given orally to neonate mice was rapidly distributed in different organs, and PTX3 ingested during lactation was detected in neonates. Finally, we observed that orally administered PTX3 provided protection against Pseudomonas aeruginosa lung infection in neonate mice. Therefore, breastfeeding constitutes, during the early life times, an important source of PTX3, which actively participates in the protection of neonates against infections. In addition, these results suggest that PTX3 might represent a therapeutic tool for treating neonatal infections and support the view that breastfeeding has beneficial effects on the neonates' health.


Asunto(s)
Lactancia Materna , Proteína C-Reactiva/fisiología , Calostro/química , Recién Nacido/inmunología , Leche Humana/química , Neumonía Bacteriana/prevención & control , Infecciones por Pseudomonas/prevención & control , Componente Amiloide P Sérico/fisiología , Administración Oral , Adulto , Animales , Animales Recién Nacidos , Mama/citología , Proteína C-Reactiva/administración & dosificación , Proteína C-Reactiva/análisis , Proteína C-Reactiva/biosíntesis , Proteína C-Reactiva/farmacocinética , Antígeno CD11b/análisis , Línea Celular , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Endotoxinas/farmacología , Endotoxinas/toxicidad , Células Epiteliales/metabolismo , Femenino , Humanos , Lactancia , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Leche Humana/citología , Monocitos/efectos de los fármacos , Monocitos/inmunología , Proteínas del Tejido Nervioso/biosíntesis , Componente Amiloide P Sérico/administración & dosificación , Componente Amiloide P Sérico/análisis , Componente Amiloide P Sérico/farmacocinética , Organismos Libres de Patógenos Específicos , Distribución Tisular
14.
Infect Immun ; 82(12): 5013-22, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25225249

RESUMEN

Murine Toll-like receptor 13 (TLR13), an endosomal receptor that is not present in humans, is activated by an unmethylated motif present in the large ribosomal subunit of bacterial RNA (23S rRNA). Little is known, however, of the impact of TLR13 on antibacterial host defenses. Here we examined the role of this receptor in the context of infection induced by the model pathogen group B streptococcus (GBS). To this end, we used bacterial strains masked from TLR13 recognition by virtue of constitutive expression of the ErmC methyltransferase, which results in dimethylation of the 23S rRNA motif at a critical adenine residue. We found that TLR13-mediated rRNA recognition was required for optimal induction of tumor necrosis factor alpha and nitrous oxide in dendritic cell and macrophage cultures stimulated with heat-killed bacteria or purified bacterial RNA. However, TLR13-dependent recognition was redundant when live bacteria were used as a stimulus. Moreover, masking bacterial rRNA from TLR13 recognition did not increase the ability of GBS to avoid host defenses and replicate in vivo. In contrast, increased susceptibility to infection was observed under conditions in which signaling by all endosomal TLRs was abolished, i.e., in mice with a loss-of-function mutation in the chaperone protein UNC93B1. Our data lend support to the conclusion that TLR13 participates in GBS recognition, although blockade of the function of this receptor can be compensated for by other endosomal TLRs. Lack of selective pressure by bacterial infections might explain the evolutionary loss of TLR13 in humans. However, further studies using different bacterial species are needed to prove this hypothesis.


Asunto(s)
Inmunidad Innata , Streptococcus agalactiae/inmunología , Receptores Toll-Like/inmunología , Animales , Células Cultivadas , Análisis por Conglomerados , ADN Bacteriano/química , ADN Bacteriano/genética , ADN Ribosómico/química , ADN Ribosómico/genética , Células Dendríticas , Macrófagos/inmunología , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Filogenia , ARN Ribosómico 23S/inmunología , Análisis de Secuencia de ADN
15.
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
16.
Front Microbiol ; 15: 1367898, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38511003

RESUMEN

The gut represents an important site of colonization of the commensal bacterium Streptococcus agalactiae (group B Streptococcus or GBS), which can also behave as a deadly pathogen in neonates and adults. Invasion of the intestinal epithelial barrier is likely a crucial step in the pathogenesis of neonatal infections caused by GBS belonging to clonal complex 17 (CC17). We have previously shown that the prototypical CC17 BM110 strain invades polarized enterocyte-like cells through their lateral surfaces using an endocytic pathway. By analyzing the cellular distribution of putative GBS receptors in human enterocyte-like Caco-2 cells, we find here that the alpha 3 (α3) and alpha 2 (α2) integrin subunits are selectively expressed on lateral enterocyte surfaces at equatorial and parabasal levels along the vertical axis of polarized cells, in an area corresponding to GBS entry sites. The α3ß1 and α2ß1 integrins were not readily accessible in fully differentiated Caco-2 monolayers but could be exposed to specific antibodies after weakening of intercellular junctions in calcium-free media. Under these conditions, anti-α3ß1 and anti-α2ß1 antibodies significantly reduced GBS adhesion to and invasion of enterocytes. After endocytosis, α3ß1 and α2ß1 integrins localized to areas of actin remodeling around GBS containing vacuoles. Taken together, these data indicate that GBS can invade enterocytes by binding to α3ß1 and α2ß1 integrins on the lateral membrane of polarized enterocytes, resulting in cytoskeletal remodeling and bacterial internalization. Blocking integrins might represent a viable strategy to prevent GBS invasion of gut epithelial tissues.

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.
Microb Pathog ; 59-60: 42-7, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23587464

RESUMEN

By screening a whole-genome λ-display library of Streptococcus pneumoniae, we have previously identified a novel surface protein, named Spr1875, that exhibited immunogenic properties and was closely related to pneumococcal virulence. In the present study, we investigated the role of the Spr1875 antigen in the interaction of S. pneumoniae with microglia, the resident brain macrophages. By using an in vitro infection model, the BV2 microglial cell line was challenged with the S. pneumoniae strain DP1004 and its isogenic spr1875-deleted mutant (Δspr1875). Both strains were phagocytosed by microglia efficiently and to a similar extent; however, the DP1004 strain was more resistant than the Δspr1875 mutant to the intracellular killing, as assessed by antibiotic protection and phagosome maturation assays. Moreover, significant differences between the two strains were also observed in terms of susceptibility to microglia-mediated killing. Taken together, these results indicate that S. pneumoniae-microglial cell interplay is influenced by the presence of Spr1875, suggesting that this protein may play a role in the pathogenesis of pneumococcal meningitis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Microglía/inmunología , Microglía/microbiología , Streptococcus pneumoniae/inmunología , Streptococcus pneumoniae/patogenicidad , Factores de Virulencia/metabolismo , Proteínas Bacterianas/genética , Línea Celular , Eliminación de Gen , Humanos , Viabilidad Microbiana , Fagocitosis , Streptococcus pneumoniae/genética , Factores de Virulencia/genética
19.
Pathogens ; 12(1)2023 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-36678467

RESUMEN

Two-component signaling systems (TCSs) are finely regulated mechanisms by which bacteria adapt to environmental conditions by modifying the expression of target genes. In bacterial pathogenesis, TCSs play important roles in modulating adhesion to mucosal surfaces, resistance to antibiotics, and metabolic adaptation. In the context of urinary tract infections (UTI), one of the most common types infections causing significant health problems worldwide, uropathogens use TCSs for adaptation, survival, and establishment of pathogenicity. For example, uropathogens can exploit TCSs to survive inside bladder epithelial cells, sense osmolar variations in urine, promote their ascension along the urinary tract or even produce lytic enzymes resulting in exfoliation of the urothelium. Despite the usefulness of studying the function of TCSs in in vitro experimental models, it is of primary necessity to study bacterial gene regulation also in the context of host niches, each displaying its own biological, chemical, and physical features. In light of this, the aim of this review is to provide a concise description of several bacterial TCSs, whose activity has been described in mouse models of UTI.

20.
Antibiotics (Basel) ; 12(6)2023 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-37370284

RESUMEN

The number of multidrug-resistant bacteria is rapidly spreading worldwide. Among the various mechanisms determining resistance to antimicrobial agents, multidrug efflux pumps play a noteworthy role because they export extraneous and noxious substrates from the inside to the outside environment of the bacterial cell contributing to multidrug resistance (MDR) and, consequently, to the failure of anti-infective therapies. The expression of multidrug efflux pumps can be under the control of transcriptional regulators and two-component systems (TCS). TCS are a major mechanism by which microorganisms sense and reply to external and/or intramembrane stimuli by coordinating the expression of genes involved not only in pathogenic pathways but also in antibiotic resistance. In this review, we describe the influence of TCS on multidrug efflux pump expression and activity in some Gram-negative and Gram-positive bacteria. Taking into account the strict correlation between TCS and multidrug efflux pumps, the development of drugs targeting TCS, alone or together with already discovered efflux pump inhibitors, may represent a beneficial strategy to contribute to the fight against growing antibiotic resistance.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA