Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 10: 2326, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31636639

RESUMEN

The ability of Helicobacter pylori to evade the host immune system allows the bacterium to colonize the host for a lifetime. Long-term infection with H. pylori causes chronic inflammation, which is the major risk factor for the development of gastric ulcers and gastric cancer. Lactobacilli are part of the human microbiota and have been studied as an adjunct treatment in H. pylori eradication therapy. However, the molecular mechanisms by which lactobacilli act against H. pylori infection have not been fully characterized. In this study, we investigated the anti-inflammatory effects of Lactobacillus strains upon coincubation of host macrophages with H. pylori. We found that Lactobacillus gasseri Kx110A1 (L. gas), a strain isolated from a human stomach, but not other tested Lactobacillus species, blocked the production of the proinflammatory cytokines TNF and IL-6 in H. pylori-infected macrophages. Interestingly, L. gas also inhibited the release of these cytokines in LPS or LTA stimulated macrophages, demonstrating a general anti-inflammatory property. The inhibition of these cytokines did not occur through the polarization of macrophages from the M1 (proinflammatory) to M2 (anti-inflammatory) phenotype or through the altered viability of H. pylori or host cells. Instead, we show that L. gas suppressed the release of TNF and IL-6 by reducing the expression of ADAM17 (also known as TNF-alpha-converting enzyme, TACE) on host cells. Our findings reveal a novel mechanism by which L. gas prevents the production of the proinflammatory cytokines TNF and IL-6 in host macrophages.


Asunto(s)
Proteína ADAM17/antagonistas & inhibidores , Citocinas/biosíntesis , Helicobacter pylori/patogenicidad , Lactobacillus gasseri/fisiología , Macrófagos/inmunología , Polaridad Celular , Humanos , Interleucina-6/biosíntesis , Interleucina-6/genética , Células THP-1 , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
2.
Eur J Immunol ; 47(1): 119-130, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27794168

RESUMEN

CD46, a membrane cofactor expressed on all nucleated human cells, plays an essential role in suppressing autoimmune reactions and protecting host cells from complement-mediated attack. Human transgenic CD46 homozygous mice (CD46+/+ ) are prone to lethal sepsis upon infection with Neisseria meningitidis (N. meningitidis). However, the underlying mechanisms are poorly understood. Here, we determined thatCD46+/+ mice produce large numbers of M1 type macrophages with enhanced surface expression of MHC II and production of pro-inflammatory mediators such as IL-6, TNF, IL-12, and IL-1ß In the presence of M-CSF or GM-CSF, CD46 signaling enhances monocyte-macrophage differentiation. Additionally, CD46+/+ macrophages rapidly undergo apoptosis upon LPS challenge or meningococcal infection, which could contribute to uncontrolled bacterial dissemination in vivo. Adoptive transfer of CD46+/+ peritoneal macrophages aggravated septic responses in wild-type mice, but the depletion of macrophages partially alleviated septic reactions in CD46+/+ mice after N. meningitidis infection. Our findings reveal a novel role of CD46 in accelerating inflammatory responses upon meningococcal infection or LPS stimulation by regulating the functional polarization and survival of macrophages.


Asunto(s)
Susceptibilidad a Enfermedades , Macrófagos/inmunología , Macrófagos/metabolismo , Proteína Cofactora de Membrana/genética , Infecciones Meningocócicas/genética , Infecciones Meningocócicas/inmunología , Neisseria meningitidis/inmunología , Sepsis , Traslado Adoptivo , Empalme Alternativo , Animales , Apoptosis/genética , Apoptosis/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunofenotipificación , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/inmunología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Proteína Cofactora de Membrana/metabolismo , Infecciones Meningocócicas/microbiología , Infecciones Meningocócicas/mortalidad , Ratones , Ratones Transgénicos , Fagocitosis/inmunología , Fenotipo , Transducción de Señal
3.
Infect Immun ; 84(5): 1501-1513, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26930706

RESUMEN

Neisseria meningitidis autoaggregation is an important step during attachment to human cells. Aggregation is mediated by type IV pili and can be modulated by accessory pilus proteins, such as PilX, and posttranslational modifications of the major pilus subunit PilE. The mechanisms underlying the regulation of aggregation remain poorly characterized. Polynucleotide phosphorylase (PNPase) is a 3'-5' exonuclease that is involved in RNA turnover and the regulation of small RNAs. In this study, we biochemically confirm that NMC0710 is the N. meningitidis PNPase, and we characterize its role in N. meningitidis pathogenesis. We show that deletion of the gene encoding PNPase leads to hyperaggregation and increased adhesion to epithelial cells. The aggregation induced was found to be dependent on pili and to be mediated by excessive pilus bundling. PNPase expression was induced following bacterial attachment to human cells. Deletion of PNPase led to global transcriptional changes and the differential regulation of 469 genes. We also demonstrate that PNPase is required for full virulence in an in vivo model of N. meningitidis infection. The present study shows that PNPase negatively affects aggregation, adhesion, and virulence in N. meningitidis.


Asunto(s)
Adhesión Bacteriana , Neisseria meningitidis/enzimología , Neisseria meningitidis/fisiología , Polirribonucleótido Nucleotidiltransferasa/metabolismo , Factores de Virulencia/metabolismo , Animales , Línea Celular , Células Epiteliales/microbiología , Eliminación de Gen , Perfilación de la Expresión Génica , Humanos , Infecciones Meningocócicas/microbiología , Infecciones Meningocócicas/patología , Ratones Transgénicos , Neisseria meningitidis/genética , Polirribonucleótido Nucleotidiltransferasa/genética , Virulencia , Factores de Virulencia/genética
4.
Infect Immun ; 84(5): 1526-1535, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26930708

RESUMEN

The human gastrointestinal tract, including the harsh environment of the stomach, harbors a large variety of bacteria, of which Lactobacillus species are prominent members. The molecular mechanisms by which species of lactobacilli interfere with pathogen colonization are not fully characterized. In this study, we aimed to study the effect of lactobacillus strains upon the initial attachment of Helicobacter pylori to host cells. Here we report a novel mechanism by which lactobacilli inhibit adherence of the gastric pathogen H. pylori In a screen with Lactobacillus isolates, we found that only a few could reduce adherence of H. pylori to gastric epithelial cells. Decreased attachment was not due to competition for space or to lactobacillus-mediated killing of the pathogen. Instead, we show that lactobacilli act on H. pylori directly by an effector molecule that is released into the medium. This effector molecule acts on H. pylori by inhibiting expression of the adhesin-encoding gene sabA Finally, we verified that inhibitory lactobacilli reduced H. pylori colonization in an in vivo model. In conclusion, certain Lactobacillus strains affect pathogen adherence by inhibiting sabA expression and thereby reducing H. pylori binding capacity.


Asunto(s)
Adhesinas Bacterianas/biosíntesis , Antibiosis , Adhesión Bacteriana , Células Epiteliales/microbiología , Helicobacter pylori/fisiología , Lactobacillus/fisiología , Animales , Línea Celular , Modelos Animales de Enfermedad , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/prevención & control , Helicobacter pylori/crecimiento & desarrollo , Humanos , Lactobacillus/crecimiento & desarrollo , Ratones Transgénicos
5.
mBio ; 7(1): e01670-15, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26884432

RESUMEN

UNLABELLED: Neisseria meningitidis colonizes the nasopharyngeal mucosa of healthy populations asymptomatically, although the bacterial surface is rich in motifs that activate the host innate immunity. What determines the tolerant host response to this bacterium in asymptomatic carriers is poorly understood. We demonstrated that the conserved meningococcal surface protein NhhA orchestrates monocyte (Mo) differentiation specifically into macrophage-like cells with a CD200R(hi) phenotype (NhhA-Mφ). In response to meningococcal stimulation, NhhA-Mφ failed to produce proinflammatory mediators. Instead, they upregulated interleukin-10 (IL-10) and Th2/regulatory T cell (Treg)-attracting chemokines, such as CCL17, CCL18, and CCL22. Moreover, NhhA-Mφ were highly efficient in eliminating bacteria. The in vivo validity of these findings was corroborated using a murine model challenged with N. meningitidis systematically or intranasally. The NhhA-modulated immune response protected mice from septic shock; Mo/Mφ depletion abolished this protective effect. Intranasal administration of NhhA induced an anti-inflammatory response, which was associated with N. meningitidis persistence at the nasopharynx. In vitro studies demonstrated that NhhA-triggered Mo differentiation occurred upon engaged Toll-like receptor 1 (TLR1)/TLR2 signaling and extracellular signal-regulated kinase (ERK) and Jun N-terminal protein kinase (JNK) activation and required endogenously produced IL-10 and tumor necrosis factor alpha (TNF-α). Our findings reveal a strategy that might be adopted by N. meningitidis to maintain asymptomatic nasopharyngeal colonization. IMPORTANCE: Neisseria meningitidis is an opportunistic human-specific pathogen that colonizes the nasopharyngeal mucosa asymptomatically in approximately 10% of individuals. Very little is known about how this bacterium evades immune activation during the carriage stage. Here, we observed that N. meningitidis, via the conserved surface protein NhhA, skewed monocyte differentiation into macrophages with a CD200R(hi) phenotype. Both in vivo and in vitro data demonstrated that these macrophages, upon meningococcal infection, played an important role in forming a homeostatic immune microenvironment through their capacity to eliminate invading bacteria and to generate anti-inflammatory mediators. This work provides novel insight into the mechanisms underlying the commensal persistence of N. meningitidis.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Portador Sano/microbiología , Homeostasis , Macrófagos/inmunología , Infecciones Meningocócicas/microbiología , Nasofaringe/microbiología , Neisseria meningitidis/fisiología , Adhesinas Bacterianas/genética , Animales , Diferenciación Celular , Quimiocina CCL17/genética , Quimiocina CCL22/genética , Quimiocinas CC/genética , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata , Interleucina-10/genética , Interleucina-10/inmunología , Infecciones Meningocócicas/inmunología , Ratones , Monocitos/inmunología , Monocitos/fisiología , Neisseria meningitidis/genética , Transducción de Señal , Simbiosis , Linfocitos T Reguladores/inmunología , Receptor Toll-Like 2/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
6.
Microbes Infect ; 18(2): 109-17, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26482500

RESUMEN

Neisseria meningitidis is an opportunistic human pathogen that usually colonizes the nasopharyngeal mucosa asymptomatically. Upon invasion into the blood and central nervous system, this bacterium triggers a fulminant inflammatory reaction with the manifestations of septicemia and meningitis, causing high morbidity and mortality. To reveal the bacterial adaptations to specific and dynamic host environments, we performed a comprehensive proteomic survey of N. meningitidis isolated from the nasal mucosa, CSF and blood of a mouse disease model. We could identify 51 proteins whose expression pattern has been changed during infection, many of which have not yet been characterized. The abundance of proteins was markedly lower in the bacteria isolated from the nasal mucosa compared to the bacteria from the blood and CSF, indicating that initiating adhesion is the harshest challenge for meningococci. The high abundance of the glutamate dehydrogenase (GdhA) and Opa1800 proteins in all bacterial isolates suggests their essential role in bacterial survival in vivo. To evaluate the biological relevance of our proteomic findings, four candidate proteins from representative functional groups, such as the bacterial chaperone GroEL, IMP dehydrogenase GuaB, and membrane proteins PilQ and NMC0101, were selected and their impact on bacterial fitness was investigated by mutagenesis assays. This study provides an integrated picture of bacterial niche-specific adaptations during consecutive infection processes.


Asunto(s)
Adaptación Fisiológica , Infecciones Meningocócicas/microbiología , Neisseria meningitidis/fisiología , Animales , Bacteriemia/microbiología , Sangre/microbiología , Portador Sano/microbiología , Líquido Cefalorraquídeo/microbiología , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Meningitis Bacterianas/microbiología , Ratones , Mucosa Nasal/microbiología , Neisseria meningitidis/química , Neisseria meningitidis/aislamiento & purificación , Proteoma/análisis , Factores de Virulencia/genética
7.
Cell Microbiol ; 17(7): 1008-20, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25600171

RESUMEN

Neisseria meningitidis, a major cause of bacterial meningitis and septicaemia, secretes multiple virulence factors, including the adhesion and penetration protein (App) and meningococcal serine protease A (MspA). Both are conserved, immunogenic, type Va autotransporters harbouring S6-family serine endopeptidase domains. Previous work suggested that both could mediate adherence to human cells, but their precise contribution to meningococcal pathogenesis was unclear. Here, we confirm that App and MspA are in vivo virulence factors since human CD46-expressing transgenic mice infected with meningococcal mutants lacking App, MspA or both had improved survival rates compared with mice infected with wild type. Confocal imaging showed that App and MspA were internalized by human cells and trafficked to the nucleus. Cross-linking and enzyme-linked immuno assay (ELISA) confirmed that mannose receptor (MR), transferrin receptor 1 (TfR1) and histones interact with MspA and App. Dendritic cell (DC) uptake could be blocked using mannan and transferrin, the specific physiological ligands for MR and TfR1, whereas in vitro clipping assays confirmed the ability of both proteins to proteolytically cleave the core histone H3. Finally, we show that App and MspA induce a dose-dependent increase in DC death via caspase-dependent apoptosis. Our data provide novel insights into the roles of App and MspA in meningococcal infection.


Asunto(s)
Apoptosis , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/metabolismo , Histonas/metabolismo , Interacciones Huésped-Patógeno , Neisseria meningitidis/patogenicidad , Sistemas de Secreción Tipo V/metabolismo , Factores de Virulencia/metabolismo , Transporte Activo de Núcleo Celular , Animales , Supervivencia Celular , Células Cultivadas , Células Dendríticas/microbiología , Células Dendríticas/fisiología , Modelos Animales de Enfermedad , Humanos , Infecciones Meningocócicas/microbiología , Infecciones Meningocócicas/patología , Ratones Transgénicos , Proteolisis , Análisis de Supervivencia
8.
PLoS One ; 9(8): e103427, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25105504

RESUMEN

Cadmium ions (Cd2+) have been reported to accumulate in bovine tissues, although Cd2+ cytotoxicity has not been investigated thoroughly in this species. Zinc ions (Zn2+) have been shown to antagonize the toxic effects of heavy metals such as Cd2+ in some systems. The present study investigated Cd2+ cytotoxicity in Madin-Darby bovine kidney (MDBK) epithelial cells, and explored whether this was modified by Zn2+. Exposure to Cd2+ led to a dose- and time-dependent increase in apoptotic cell death, with increased intracellular levels of reactive oxygen species and mitochondrial damage. Zn2+ supplementation alleviated Cd2+-induced cytotoxicity and this protective effect was more obvious when cells were exposed to a lower concentration of Cd2+ (10 µM), as compared to 50 µM Cd2+. This indicated that high levels of Cd2+ accumulation might induce irreversible damage in bovine kidney cells. Metallothioneins (MTs) are metal-binding proteins that play an essential role in heavy metal ion detoxification. We found that co-exposure to Zn2+ and Cd2+ synergistically enhanced RNA and protein expression of MT-1, MT-2, and the metal-regulatory transcription factor 1 in MDBK cells. Notably, addition of Zn2+ reduced the amounts of cytosolic Cd2+ detected following MDBK exposure to 10 µM Cd2+. These findings revealed a protective role of Zn2+ in counteracting Cd2+ uptake and toxicity in MDBK cells, indicating that this approach may provide a means to protect livestock from excessive Cd2+ accumulation.


Asunto(s)
Cadmio/farmacocinética , Cadmio/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Zinc/farmacología , Animales , Apoptosis/efectos de los fármacos , Bovinos , Línea Celular , Suplementos Dietéticos , Relación Dosis-Respuesta a Droga , Hepatocitos/metabolismo , Metalotioneína/metabolismo , Factores de Tiempo , Zinc/administración & dosificación
9.
Open Biol ; 3(10): 130048, 2013 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-24107297

RESUMEN

Interactions between commensal pathogens and hosts are critical for disease development but the underlying mechanisms for switching between the commensal and virulent states are unknown. We show that the human pathogen Neisseria meningitidis, the leading cause of pyogenic meningitis, can modulate gene expression via uptake of host pro-inflammatory cytokines leading to increased virulence. This uptake is mediated by type IV pili (Tfp) and reliant on the PilT ATPase activity. Two Tfp subunits, PilE and PilQ, are identified as the ligands for TNF-α and IL-8 in a glycan-dependent manner, and their deletion results in decreased virulence and increased survival in a mouse model. We propose a novel mechanism by which pathogens use the twitching motility mode of the Tfp machinery for sensing and importing host elicitors, aligning with the inflamed environment and switching to the virulent state.


Asunto(s)
Citocinas/metabolismo , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/metabolismo , Interleucina-8/metabolismo , Meningitis Bacterianas/microbiología , Neisseria meningitidis/patogenicidad , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Proteínas Fimbrias/genética , Fimbrias Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano , Humanos , Ligandos , Meningitis Bacterianas/metabolismo , Ratones , Ratones Transgénicos , Neisseria meningitidis/genética , Neisseria meningitidis/metabolismo , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
10.
Antimicrob Agents Chemother ; 57(8): 3704-12, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23689723

RESUMEN

Meningococcal disease is characterized by a fast progression and a high mortality rate. Cell-penetrating peptides (CPPs), developed as vectors for cargo delivery into eukaryotic cells, share structural features with antimicrobial peptides. A screen identified two CPPs, transportan-10 (TP10) and model amphipathic peptide (MAP), with bactericidal action against Neisseria meningitidis. Both peptides were active in human whole blood at micromolar concentrations, while hemolysis remained negligible. Additionally, TP10 exhibited significant antibacterial activity in vivo. Uptake of SYTOX green into live meningococci was observed within minutes after TP10 treatment, suggesting that TP10 may act by membrane permeabilization. Apart from its bactericidal activity, TP10 suppressed inflammatory cytokine release from macrophages infected with N. meningitidis as well as from macrophages stimulated with enterobacterial and meningococcal lipopolysaccharide (LPS). Finally, incubation with TP10 reduced the binding of LPS to macrophages. This novel endotoxin-inhibiting property of TP10, together with its antimicrobial activity in vivo, indicates the possibility to design peptide-based therapies for infectious diseases.


Asunto(s)
Péptidos de Penetración Celular/aislamiento & purificación , Péptidos de Penetración Celular/farmacología , Galanina/farmacología , Inflamación/tratamiento farmacológico , Neisseria meningitidis/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Venenos de Avispas/farmacología , Animales , Antibacterianos/síntesis química , Antibacterianos/farmacología , Antiinflamatorios/inmunología , Antiinflamatorios/farmacología , Péptidos Catiónicos Antimicrobianos/síntesis química , Péptidos Catiónicos Antimicrobianos/aislamiento & purificación , Péptidos Catiónicos Antimicrobianos/farmacología , Membrana Celular , Péptidos de Penetración Celular/síntesis química , Citocinas/inmunología , Evaluación Preclínica de Medicamentos , Galanina/inmunología , Humanos , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Proteína Cofactora de Membrana/genética , Proteína Cofactora de Membrana/metabolismo , Infecciones Meningocócicas/tratamiento farmacológico , Ratones , Ratones Transgénicos , Proteínas Recombinantes de Fusión/inmunología , Venenos de Avispas/inmunología
11.
Infect Immun ; 80(11): 4027-33, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22949555

RESUMEN

Activation of macrophages by Toll-like receptors (TLRs) and functionally related proteins is essential for host defense and innate immunity. TLRs recognize a wide variety of pathogen-associated molecules. Here, we demonstrate that the meningococcal outer membrane protein NhhA has immunostimulatory functions and triggers release of proinflammatory cytokines from macrophages. NhhA-induced cytokine release was found to proceed via two distinct pathways in RAW 264.7 macrophages. Interleukin-6 (IL-6) secretion was dependent on activation of TLR4 and required the TLR signaling adaptor protein MyD88. In contrast, release of tumor necrosis factor (TNF) was TLR4 and MyD88 independent. Both pathways involved NF-κB-dependent gene regulation. Using a PCR-based screen, we could identify additional targets of NhhA-dependent gene activation such as the cytokines and growth factors IL-1α, IL-1ß, granulocyte colony-stimulating factor (G-CSF), and granulocyte-macrophage colony-stimulating factor (GM-CSF). In human monocyte-derived macrophages, G-CSF, GM-CSF, and IL-6 were found to be major targets of NhhA-dependent gene regulation. NhhA induced transcription of IL-6 and G-CSF mRNA via TLR4-dependent pathways, whereas GM-CSF transcription was induced via TLR4-independent pathways. These data provide new insights into the role of NhhA in host-pathogen interaction.


Asunto(s)
Adhesinas Bacterianas/fisiología , Citocinas/biosíntesis , Macrófagos/inmunología , Glicoproteínas de Membrana/inmunología , FN-kappa B/metabolismo , Neisseria meningitidis/inmunología , Receptor Toll-Like 4/genética , Células Cultivadas , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Interleucina-6/biosíntesis , Macrófagos/metabolismo , Glicoproteínas de Membrana/genética , Monocitos/inmunología , Transducción de Señal , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo
12.
PLoS One ; 7(7): e41445, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22844479

RESUMEN

Euthyroid sick syndrome characterized by reduced levels of thyroid hormones (THs) is observed in patients with meningococcal shock. It has been found that the level of THs reflects disease severity and is predictive for mortality. The present study was conducted to investigate the impact of THs on host defense during meningococcal infection. We found that supplementation of thyroxine to mice infected with Neisseria meningitidis enhanced bacterial clearance, attenuated the inflammatory responses and promoted survival. In vitro studies with macrophages revealed that THs enhanced bacteria-cell interaction and intracellular killing of meningococci by stimulating inducible nitric oxide synthase (iNos)-mediated NO production. TH treatment did not activate expression of TH receptors in macrophages. Instead, the observed TH-directed actions were mediated through nongenomic pathways involving the protein kinases PI3K and ERK1/2 and initiated at the membrane receptor integrin αvß3. Inhibition of nongenomic TH signaling prevented iNos induction, NO production and subsequent intracellular bacterial killing by macrophages. These data demonstrate a beneficial role of THs in macrophage-mediated N. meningitidis clearance. TH replacement might be a novel option to control meningococcal septicemia.


Asunto(s)
Meningitis Meningocócica/metabolismo , Meningitis Meningocócica/microbiología , Neisseria meningitidis/efectos de los fármacos , Neisseria meningitidis/patogenicidad , Óxido Nítrico/metabolismo , Hormonas Tiroideas/farmacología , Animales , Línea Celular , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Integrina alfaVbeta3/metabolismo , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Espacio Intracelular/microbiología , Meningitis Meningocócica/patología , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia
13.
PLoS One ; 7(1): e29586, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22238624

RESUMEN

Phagocytotic cells play a fundamental role in the defense against bacterial pathogens. One mechanism whereby bacteria evade phagocytosis is to produce factors that trigger apoptosis. Here we identify for the first time a meningococcal protein capable of inducing macrophage apoptosis. The conserved meningococcal outer membrane protein NhhA (Neisseria hia/hsf homologue A, also known as Hsf) mediates bacterial adhesion and interacts with extracellular matrix components heparan sulphate and laminin. Meningococci lacking NhhA fail to colonise nasal mucosa in a mouse model of meningococcal disease. We found that exposure of macrophages to NhhA resulted in a highly increased rate of apoptosis that proceeded through caspase activation. Exposure of macrophages to NhhA also led to iNOS induction and nitric oxide production. However, neither nitric oxide production nor TNF-α signaling was found to be a prerequisite for NhhA-induced apoptosis. Macrophages exposed to wildtype NhhA-expressing meningococci were also found to undergo apoptosis whereas NhhA-deficient meningococci had a markedly decreased capacity to induce macrophage apoptosis. These data provide new insights on the role of NhhA in meningococcal disease. NhhA-induced macrophage apoptosis could be a mechanism whereby meningococci evade immunoregulatory and phagocytotic actions of macrophages.


Asunto(s)
Apoptosis , Proteínas de la Membrana Bacteriana Externa/fisiología , Macrófagos/fisiología , Infecciones Meningocócicas/patología , Neisseria meningitidis/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Apoptosis/fisiología , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas de la Membrana Bacteriana Externa/farmacología , Linaje de la Célula , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Infecciones Meningocócicas/inmunología , Ratones , Monocitos/citología , Monocitos/fisiología , Neisseria meningitidis/genética , Óxido Nítrico/metabolismo , Óxido Nítrico/fisiología , Unión Proteica , Transducción de Señal/genética , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
14.
Methods Mol Biol ; 799: 153-68, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21993645

RESUMEN

Neisseria meningitidis is a human specific organism that causes severe sepsis and/or meningitis with high mortality. The disease scenario is rapid and much remains unknown about the disease process and host-pathogen interaction. In this chapter, we describe a protocol for generating a bioluminescently labeled N. meningitidis strain in order to advance our understanding of meningococcal disease progression. We also describe how in vivo bioluminescence imaging (BLI) can be used to observe novel features of the disease dynamics during meningococcal infection.


Asunto(s)
Sustancias Luminiscentes/metabolismo , Mediciones Luminiscentes/métodos , Infecciones Meningocócicas/diagnóstico , Infecciones Meningocócicas/fisiopatología , Modelos Animales , Neisseria meningitidis , Animales , Southern Blotting , Clonación Molecular/métodos , Cartilla de ADN/genética , Electroforesis en Gel de Agar , Humanos , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa
15.
PLoS One ; 6(7): e21749, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21747953

RESUMEN

Bacterial auto-aggregation is a critical step during adhesion of N. meningitidis to host cells. The precise mechanisms and functions of bacterial auto-aggregation still remain to be fully elucidated. In this work, we characterize the role of a meningococcal hypothetical protein, NMB0995/NMC0982, and show that this protein, here denoted NafA, acts as an anti-aggregation factor. NafA was confirmed to be surface exposed and was found to be induced at a late stage of bacterial adherence to epithelial cells. A NafA deficient mutant was hyperpiliated and formed bundles of pili. Further, the mutant displayed increased adherence to epithelial cells when compared to the wild-type strain. In the absence of host cells, the NafA deficient mutant was more aggregative than the wild-type strain. The in vivo role of NafA in sepsis was studied in a murine model of meningococcal disease. Challenge with the NafA deficient mutant resulted in lower bacteremia levels and mortality when compared to the wild-type strain. The present study reveals that meningococcal NafA is an anti-aggregation factor with strong impact on the disease outcome. These data also suggest that appropriate bacterial auto-aggregation is controlled by both aggregation and anti-aggregation factors during Neisseria infection in vivo.


Asunto(s)
Proteínas Bacterianas/metabolismo , Fimbrias Bacterianas/metabolismo , Neisseria meningitidis/citología , Neisseria meningitidis/metabolismo , Secuencia de Aminoácidos , Animales , Adhesión Bacteriana , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Secuencia Conservada , Células Epiteliales/citología , Células Epiteliales/microbiología , Humanos , Infecciones Meningocócicas/sangre , Infecciones Meningocócicas/microbiología , Ratones , Datos de Secuencia Molecular , Peso Molecular , Neisseria meningitidis/patogenicidad , Regulación hacia Arriba
16.
Gastroenterology ; 141(3): 918-28, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21699774

RESUMEN

BACKGROUND & AIMS: CD46 is a C3b/C4b binding complement regulator and a receptor for several human pathogens. We examined the interaction between CD46 and Helicobacter pylori (a bacterium that colonizes the human gastric mucosa and causes gastritis), peptic ulcers, and cancer. METHODS: Using gastric epithelial cells, we analyzed a set of H pylori strains and mutants for their ability to interact with CD46 and/or influence CD46 expression. Bacterial interaction with full-length CD46 and small CD46 peptides was evaluated by flow cytometry, fluorescence microscopy, enzyme-linked immunosorbent assay, and bacterial survival analyses. RESULTS: H pylori infection caused shedding of CD46 into the extracellular environment. A soluble form of CD46 bound to H pylori and inhibited growth, in a dose- and time-dependent manner, by interacting with urease and alkyl hydroperoxide reductase, which are essential bacterial pathogenicity-associated factors. Binding of CD46 or CD46-derived synthetic peptides blocked the urease activity and ability of bacteria to survive in acidic environments. Oral administration of one CD46 peptide eradicated H pylori from infected mice. CONCLUSIONS: CD46 is an antimicrobial agent that can eradicate H pylori. CD46 peptides might be developed to treat H pylori infection.


Asunto(s)
Antibacterianos/farmacología , Mucosa Gástrica/metabolismo , Helicobacter pylori/efectos de los fármacos , Proteína Cofactora de Membrana/farmacología , Ureasa/efectos de los fármacos , Ureasa/metabolismo , Animales , Antibacterianos/uso terapéutico , Línea Celular , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Mucosa Gástrica/citología , Mucosa Gástrica/microbiología , Infecciones por Helicobacter/tratamiento farmacológico , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/crecimiento & desarrollo , Helicobacter pylori/metabolismo , Humanos , Proteína Cofactora de Membrana/metabolismo , Proteína Cofactora de Membrana/uso terapéutico , Ratones , Ratones Mutantes , Peroxirredoxinas/efectos de los fármacos , Peroxirredoxinas/metabolismo , Factores de Tiempo , Resultado del Tratamiento
17.
PLoS One ; 5(11): e14034, 2010 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21124975

RESUMEN

Neisseria meningitidis is a human-specific pathogen with capacity to cause septic shock and meningitis. It has been hypothesized that invasion of the central nervous system (CNS) is a complication of a bacteremic condition. In this study, we aimed to characterize the invasion route of N. meningitidis to the CNS. Using an intranasally challenged mouse disease model, we found that twenty percent of the mice developed lethal meningitis even though no bacteria could be detected in blood. Upon bacterial infection, epithelial lesions and redistribution of intracellular junction protein N-cadherin were observed at the nasal epithelial mucosa, especially at the olfactory epithelium, which is functionally and anatomically connected to the CNS. Bacteria were detected in the submucosa of the olfactory epithelium, along olfactory nerves in the cribriform plate, at the olfactory bulb and subsequently at the meninges and subarachnoid space. Furthermore, our data suggest that a threshold level of bacteremia is required for the development of meningococcal sepsis. Taken together, N. meningitidis is able to pass directly from nasopharynx to meninges through the olfactory nerve system. This study enhances our understanding how N. meningitidis invades the meninges. The nasal olfactory nerve system may be a novel target for disease prevention that can improve outcome and survival.


Asunto(s)
Meninges/microbiología , Meningitis Meningocócica/microbiología , Neisseria meningitidis/fisiología , Nervio Olfatorio/microbiología , Animales , Bacteriemia/microbiología , Cadherinas/metabolismo , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/microbiología , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Patógeno , Humanos , Masculino , Proteína Cofactora de Membrana/genética , Proteína Cofactora de Membrana/metabolismo , Meningitis Meningocócica/sangre , Meningitis Meningocócica/líquido cefalorraquídeo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Lavado Nasal (Proceso) , Mucosa Nasal/metabolismo , Mucosa Nasal/microbiología , Mucosa Olfatoria/metabolismo , Mucosa Olfatoria/microbiología , Nervio Olfatorio/metabolismo
18.
Microbes Infect ; 10(12-13): 1325-34, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18761100

RESUMEN

Lactobacilli are normal inhabitants of our microbiota and are known to protect against pathogens. Neisseria gonorrhoeae is a human specific pathogenic bacterium that colonises the urogenital tract where it causes gonorrhoea. In this study we analysed early interactions between lactobacilli and gonococci and investigated how they compete for adherence to human epithelial cervical cells. We show that lactobacilli adhere at various levels and that the number of adherent bacteria does not correlate to the level of protection against gonococcal infection. Protection against gonococcal adhesion varied between Lactobacillus species. Lactobacillus crispatus, Lactobacillus gasseri and Lactobacillus reuteri were capable of reducing gonococcal adherence while Lactobacillus rhamnosus was not. Lactobacillus strains of vaginal origin had the best capacity to remain attached to the host cell during gonococcal adherence. Further, we show that gonococci and lactobacilli interact with each other with resultant lactobacilli incorporation into the gonococcal microcolony. Hence, gonococci bind to colonised lactobacilli and this complex frequently detaches from the epithelial cell surface, resulting in reduced bacterial colonisation. Also, purified gonococcal pili are capable of removing adherent lactobacilli from the cell surface. Taken together, we reveal novel data regarding gonococcal and lactobacilli competition for adherence that will benefit future gonococcal prevention and treatments.


Asunto(s)
Adhesión Bacteriana , Cuello del Útero/microbiología , Células Epiteliales/microbiología , Lactobacillus/fisiología , Neisseria gonorrhoeae/fisiología , Línea Celular Tumoral , Cuello del Útero/citología , Técnicas de Cocultivo , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Lactobacillus/clasificación , Lactobacillus/aislamiento & purificación , Lactobacillus/patogenicidad , Microscopía Fluorescente , Neisseria gonorrhoeae/patogenicidad
19.
Infect Immun ; 76(11): 5412-20, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18794285

RESUMEN

Neisseria meningitidis is a leading cause of meningitis and septicemia worldwide, with a rapid onset of disease and a high morbidity and mortality. NhhA is a meningococcal outer membrane protein included in the family of trimeric autotransporter adhesins. The protein binds to the extracellular matrix proteins heparan sulfate and laminin and facilitates attachment to host epithelial cells. In this study, we show that NhhA is essential for bacterial colonization of the nasopharyngeal mucosa in a murine model of meningococcal disease. Successful colonization depends on bacterial attachment but also to the capacity to overcome innate host immune responses. We found that NhhA protected bacteria from phagocytosis, which is important for the mucosal survival of bacteria. In addition, NhhA mediated extensive serum resistance that increased bacterial survival in blood and promoted lethal sepsis. The presence of NhhA protected bacteria from complement-mediated killing by preventing the deposition of the membrane attack complex. Taken together, the results of this work reveal that NhhA inhibits phagocytosis and protects bacteria against complement-mediated killing, which enhances both nasal colonization and the development of sepsis in vivo.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Proteínas del Sistema Complemento/fisiología , Infecciones Meningocócicas/metabolismo , Neisseria meningitidis/patogenicidad , Fagocitosis/fisiología , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/inmunología , Animales , Adhesión Bacteriana/fisiología , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Infecciones Meningocócicas/genética , Infecciones Meningocócicas/inmunología , Ratones , Ratones Transgénicos , Mucosa Nasal/microbiología , Neisseria meningitidis/genética , Neisseria meningitidis/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Infect Immun ; 76(11): 5421-8, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18794294

RESUMEN

Neisseria meningitidis is a leading cause of meningitis and sepsis. The pathogenesis of meningococcal disease is determined by both bacterial virulence factors and the host inflammatory response. Toll-like receptors (TLRs) are prominent activators of the inflammatory response, and TLR2, -4, and -9 have been reported to be involved in the host response to N. meningitidis. While TLR4 has been suggested to play an important role in early containment of infection, the roles of TLR2 and TLR9 in meningococcal disease are not well described. Using a model for meningococcal sepsis, we report that TLR9(-/-) mice displayed reduced survival and elevated levels of bacteremia compared to wild-type mice. In contrast, TLR2(-/-) mice controlled the infection in a manner comparable to that of wild-type mice. TLR9 deficiency was also associated with reduced bactericidal activity in vitro, which was accompanied by reduced production of nitric oxide by TLR9-deficient macrophages. Interestingly, TLR9(-/-) mice recruited more macrophages to the bloodstream than wild-type mice and produced elevated levels of cytokines at late time points during infection. At the cellular level, activation of signal transduction and induction of cytokine gene expression were independent of TLR2 or TLR9 in macrophages and conventional dendritic cells. In contrast, plasmacytoid dendritic cells relied entirely on TLR9 to induce these activities. Thus, our data demonstrate an important role for TLR9 in host defense against N. meningitidis.


Asunto(s)
Bacteriemia/inmunología , Infecciones Meningocócicas/inmunología , Neisseria meningitidis/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 9/inmunología , Animales , Bacteriemia/metabolismo , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Expresión Génica/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Infecciones Meningocócicas/metabolismo , Ratones , Ratones Noqueados , Neisseria meningitidis/metabolismo , Óxido Nítrico/biosíntesis , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 9/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...