Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 18(5): 509-518, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28319098

RESUMEN

The retention of intracellular Toll-like receptors (TLRs) in the endoplasmic reticulum prevents their activation under basal conditions. TLR9 is activated by sensing ligands in specific endosomal-lysosomal compartments. Here we identified IRAP+ endosomes as major cellular compartments for the early steps of TLR9 activation in dendritic cells (DCs). Both TLR9 and its ligand, the dinucleotide CpG, were present as cargo in IRAP+ endosomes. In the absence of the aminopeptidase IRAP, the trafficking of CpG and TLR9 to lysosomes and signaling via TLR9 were enhanced in DCs and in mice following bacterial infection. IRAP stabilized CpG-containing endosomes by interacting with the actin-nucleation factor FHOD4, which slowed the trafficking of TLR9 toward lysosomes. Thus, endosomal retention of TLR9 via the interaction of IRAP with the actin cytoskeleton is a mechanism that prevents hyper-activation of TLR9 in DCs.


Asunto(s)
Cistinil Aminopeptidasa/metabolismo , Citoesqueleto/metabolismo , Células Dendríticas/fisiología , Endosomas/metabolismo , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Receptor Toll-Like 9/metabolismo , Animales , Células Cultivadas , Islas de CpG/genética , Cistinil Aminopeptidasa/genética , Células Dendríticas/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Oligodesoxirribonucleótidos/inmunología , Unión Proteica , Transducción de Señal
2.
J Infect Dis ; 221(6): 1000-1005, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-31678998

RESUMEN

Human bronchial epithelial cells play a key role in airway immune homeostasis. We hypothesized that these sentinel cells can remember a previous contact with pathogen compounds and respond nonspecifically to reinfection, a phenomenon called innate immune memory. We demonstrated that their preexposure to Pseudomonas aeruginosa flagellin modify their inflammatory response to a second, nonrelated stimulus, including live pathogens or lipopolysaccharide. Using histone acetyltransferase and methyltransferase inhibitors, we showed that this phenomenon relied on epigenetic regulation. This report is a major breakthrough in the field of multimicrobial respiratory tract infections, wherein control of inflammatory exacerbations is a major therapeutic issue.


Asunto(s)
Memoria Inmunológica , Mucosa Respiratoria/citología , Epigénesis Genética , Células Epiteliales/inmunología , Flagelina/inmunología , Regulación de la Expresión Génica/inmunología , Humanos , Inflamación , Lipopolisacáridos , Prueba de Estudio Conceptual , Pseudomonas aeruginosa/inmunología , ARN Mensajero , Mucosa Respiratoria/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
3.
J Immunol ; 194(11): 5312-9, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25904549

RESUMEN

Staphylococcus aureus is a common cause of bacterial infections in respiratory diseases. It secretes molecules to dampen host immunity, and the recently identified adenosine is one of these molecules. The type IIA secretory phospholipase A2 (sPLA2-IIA) is a host protein endowed with antibacterial properties, especially against Gram-positive bacteria such as S. aureus. However, the role of adenosine in sPLA2-IIA-mediated S. aureus killing by host is still unknown. The present studies showed that the S. aureus mutant lacking adenosine production (∆adsA strain) increased sPLA2-IIA expression in guinea pig airways and was cleared more efficiently, compared with the wild-type strain. S. aureus ∆adsA strain induced sPLA2-IIA expression by alveolar macrophages after phagocytic process via NOD2-NF-κB-dependent mechanism. However, S. aureus adenosine (wild-type and adsA-complemented strains) and exogenous adenosine downregulated S. aureus phagocytosis by alveolar macrophages, leading to inhibition of sPLA2-IIA expression. This occurred through inhibition of p38 phosphorylation via adenosine receptors A2a-, A2b-, and protein kinase A-dependent pathways. Taken together, our studies suggest that, in the airway, S. aureus escapes sPLA2-IIA-mediated killing through adenosine-mediated inhibition of phagocytosis and sPLA2-IIA expression.


Asunto(s)
Adenosina/inmunología , Fosfolipasas A2 Grupo II/biosíntesis , Interacciones Huésped-Patógeno , Macrófagos Alveolares/inmunología , Fagocitosis/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Adenosina/genética , Animales , Líquido del Lavado Bronquioalveolar , Células CHO , Línea Celular , Cricetinae , Cricetulus , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fosfolipasas A2 Grupo II/antagonistas & inhibidores , Cobayas , Imidazoles/farmacología , Masculino , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD2/inmunología , Fosforilación , Piridinas/farmacología , Receptor de Adenosina A2A/metabolismo , Receptor de Adenosina A2B/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
4.
Am J Physiol Lung Cell Mol Physiol ; 310(7): L689-99, 2016 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-26747784

RESUMEN

Influenza A viruses (IAV) trigger contagious acute respiratory diseases. A better understanding of the molecular mechanisms of IAV pathogenesis and host immune responses is required for the development of more efficient treatments of severe influenza. Calpains are intracellular proteases that participate in diverse cellular responses, including inflammation. Here, we used in vitro and in vivo approaches to investigate the role of calpain signaling in IAV pathogenesis. Calpain expression and activity were found altered in IAV-infected bronchial epithelial cells. With the use of small-interfering RNA (siRNA) gene silencing, specific synthetic inhibitors of calpains, and mice overexpressing calpastatin, we found that calpain inhibition dampens IAV replication and IAV-triggered secretion of proinflammatory mediators and leukocyte infiltration. Remarkably, calpain inhibition has a protective impact in IAV infection, since it significantly reduced mortality of mice challenged not only by seasonal H3N2- but also by hypervirulent H5N1 IAV strains. Hence, our study suggests that calpains are promising therapeutic targets for treating IAV acute pneumonia.


Asunto(s)
Calpaína/fisiología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Gripe Humana/enzimología , Animales , Citocinas/metabolismo , Perros , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Viral de la Expresión Génica , Humanos , Gripe Humana/inmunología , Gripe Humana/virología , Células de Riñón Canino Madin Darby , Ratones Endogámicos C57BL , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal , Replicación Viral
5.
PLoS Pathog ; 9(11): e1003716, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244155

RESUMEN

α-(1,3)-Glucan is a major component of the cell wall of Aspergillus fumigatus, an opportunistic human fungal pathogen. There are three genes (AGS1, AGS2 and AGS3) controlling the biosynthesis of α-(1,3)-glucan in this fungal species. Deletion of all the three AGS genes resulted in a triple mutant that was devoid of α-(1,3)-glucan in its cell wall; however, its growth and germination was identical to that of the parental strain in vitro. In the experimental murine aspergillosis model, this mutant was less pathogenic than the parental strain. The AGS deletion resulted in an extensive structural modification of the conidial cell wall, especially conidial surface where the rodlet layer was covered by an amorphous glycoprotein matrix. This surface modification was responsible for viability reduction of conidia in vivo, which explains decrease in the virulence of triple agsΔ mutant.


Asunto(s)
Aspergilosis/enzimología , Aspergillus fumigatus/enzimología , Aspergillus fumigatus/patogenicidad , Pared Celular/enzimología , Polisacáridos Fúngicos/biosíntesis , Proteínas Fúngicas/metabolismo , Glucosiltransferasas/metabolismo , Animales , Aspergilosis/genética , Aspergilosis/patología , Aspergillus fumigatus/genética , Pared Celular/genética , Polisacáridos Fúngicos/genética , Proteínas Fúngicas/genética , Eliminación de Gen , Glucosiltransferasas/genética , Humanos , Ratones Noqueados , Esporas Fúngicas/enzimología , Esporas Fúngicas/genética
6.
Cell Microbiol ; 16(10): 1503-17, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24779433

RESUMEN

Uropathogenic Escherichia coli (UPEC) colonizing kidneys is the main cause of acute pyelonephritis. TLR5 that senses flagellin was shown to be highly expressed in the bladder and to participate in host defence against flagellated UPEC, although its role in kidneys still remains elusive. Here we show that TLR5 is expressed in renal medullary collecting duct (MCD) cells, which represent a preferential site of UPEC adhesion. Flagellin, like lipopolysaccharide, stimulated the production of the chemoattractant chemokines CXCL1 and CXCL2, and subsequent migration capacity of neutrophils in cultured wild-type (WT) and Tlr4(-/-) MCDs, but not in Tlr5(-/-) MCDs. UPEC can translocate across intact MCD layers without altering tight junctions. Strikingly, the invasion capacity and transcellular translocation of the UPEC strain HT7 were significantly lower in Tlr5(-/-) than in WT MCDs. The non-motile HT7ΔfliC mutant lacking flagellin also exhibited much lower translocation capacities than the HT7 isolates. Finally, Tlr5(-/-) kidneys exhibited less infiltrating neutrophils than WT kidneys one day after the transurethral inoculation of HT7, and greater delayed renal bacterial loads in the day 4 post-infected Tlr5(-/-) kidneys. Overall, these findings indicate that the epithelial TLR5 participates to renal antibacterial defence, but paradoxically favours the translocation of UPEC across intact MCD cell layers.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Flagelina/inmunología , Túbulos Renales Colectores/inmunología , Receptor Toll-Like 5/inmunología , Escherichia coli Uropatógena/patogenicidad , Animales , Adhesión Bacteriana/fisiología , Carga Bacteriana/inmunología , Quimiocina CXCL1/biosíntesis , Quimiocina CXCL2/biosíntesis , Infecciones por Escherichia coli/microbiología , Femenino , Túbulos Renales Colectores/citología , Túbulos Renales Colectores/metabolismo , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/genética , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Pielonefritis/inmunología , Pielonefritis/microbiología , Transducción de Señal , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 5/genética , Vejiga Urinaria/inmunología , Vejiga Urinaria/metabolismo , Infecciones Urinarias/inmunología , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena/inmunología
7.
Am J Respir Crit Care Med ; 189(7): 799-811, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24555512

RESUMEN

RATIONALE: Pseudomonas aeruginosa, a major problem pathogen responsible for severe infections in critically ill patients, triggers, through a functional type-3 secretion system (T3SS), the activation of an intracellular cytosolic sensor of innate immunity, NLRC4. Although the NLRC4-inflammasome-dependent response contributes to increased clearance of intracellular pathogens, it seems that NLRC4 inflammasome activation decreases the clearance of P. aeruginosa, a mainly extracellular pathogen. OBJECTIVES: We sought to determine the underlying mechanisms of this effect of the activation of NLRC4 by P. aeruginosa. METHODS: We established acute lung injury in wild-type and Nlrc4(-/-) mice using sublethal intranasal inocula of P. aeruginosa strain CHA expressing or not a functional T3SS. We studied 96-hour survival, lung injury, bacterial clearance from the lungs, cytokine secretion in bronchoalveolar lavage, lung antimicrobial peptide expression by quantitative polymerase chain reaction, and flow cytometry analysis of lung cells. MEASUREMENTS AND MAIN RESULTS: Nlrc4(-/-) mice showed enhanced bacterial clearance and decreased lung injury contributing to increased survival against extracellular P. aeruginosa strain expressing a functional T3SS. The mechanism involved decreased NLRC4-inflammasome-driven IL-18 secretion attenuating lung injury caused by excessive neutrophil recruitment. Additionally, in the lungs of Nlrc4(-/-) mice secretion of IL-17 by innate immune cells was increased and responsible for increased expression of lung epithelial antimicrobial peptides. Furthermore, IL-18 secretion was found to repress IL-17 and IL-17-driven lung antimicrobial peptide expression. CONCLUSIONS: We report a new role of the T3SS apparatus itself, independently of exotoxin translocation. Through NLRC4 inflammasome activation, the T3SS promotes IL-18 secretion, which dampens a beneficial IL-17-mediated antimicrobial host response.


Asunto(s)
Lesión Pulmonar Aguda/microbiología , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/metabolismo , Inflamasomas/metabolismo , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/patogenicidad , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/metabolismo , Animales , Biomarcadores/metabolismo , Líquido del Lavado Bronquioalveolar/química , Caspasa 1/metabolismo , Células Cultivadas , Femenino , Citometría de Flujo , Inmunidad Innata , Interleucina-17/metabolismo , Interleucina-18/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Proc Natl Acad Sci U S A ; 109(5): 1619-24, 2012 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22307620

RESUMEN

A deficit in early clearance of Pseudomonas aeruginosa (P. aeruginosa) is crucial in nosocomial pneumonia and in chronic lung infections. Few studies have addressed the role of Toll-like receptors (TLRs), which are early pathogen associated molecular pattern receptors, in pathogen uptake and clearance by alveolar macrophages (AMs). Here, we report that TLR5 engagement is crucial for bacterial clearance by AMs in vitro and in vivo because unflagellated P. aeruginosa or different mutants defective in TLR5 activation were resistant to AM phagocytosis and killing. In addition, the clearance of PAK (a wild-type P. aeruginosa strain) by primary AMs was causally associated with increased IL-1ß release, which was dramatically reduced with PAK mutants or in WT PAK-infected primary TLR5(-/-) AMs, demonstrating the dependence of IL-1ß production on TLR5. We showed that this IL-1ß production was important in endosomal pH acidification and in inducing the killing of bacteria by AMs through asparagine endopeptidase (AEP), a key endosomal cysteine protease. In agreement, AMs from IL-1R1(-/-) and AEP(-/-) mice were unable to kill P. aeruginosa. Altogether, these findings demonstrate that TLR5 engagement plays a major role in P. aeruginosa internalization and in triggering IL-1ß formation.


Asunto(s)
Endopeptidasas/metabolismo , Interleucina-1beta/metabolismo , Macrófagos Alveolares/inmunología , Fagocitosis , Pseudomonas aeruginosa/inmunología , Receptor Toll-Like 5/fisiología , Animales , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Endogámicos C57BL
9.
J Infect Dis ; 210(2): 214-23, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24493823

RESUMEN

Influenza A virus triggers a contagious respiratory disease that can cause considerable morbidity and mortality. Using an in vitro approach, we previously demonstrated that the pattern recognition receptor retinoic acid-inducible gene I (RIG-I) plays a key role in influenza A virus-mediated immune response. However, the importance of RIG-I signaling in vivo has not been thoroughly examined, because of the lack of an appropriate mouse models. To circumvent this issue, we generated a new transgenic mouse overexpressing LGP2 (hereafter, "LGP2 TG mice"), a major regulator of the RIG-I signaling pathway. The time course of several parameters was compared in infected wild-type and LGP2 TG mice. We found that LGP2 TG mice displayed significantly reduced inflammatory mediators and a lower leukocyte infiltration into the bronchoalveolar airspace. More importantly, LGP2 TG mice had a significant survival advantage. Hence, our in vivo study reveals that LGP2 is a major downregulator of the influenza A virus-triggered detrimental inflammatory response.


Asunto(s)
Interacciones Huésped-Patógeno , Virus de la Influenza A/fisiología , ARN Helicasas/metabolismo , Animales , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Mediadores de Inflamación/análisis , Leucocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Transducción de Señal , Análisis de Supervivencia
10.
PLoS Pathog ; 8(8): e1002841, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22916010

RESUMEN

Intracellular Toll-like receptors (TLRs) expressed by dendritic cells recognize nucleic acids derived from pathogens and play an important role in the immune responses against the influenza virus (IAV), a single-stranded RNA sensed by different receptors including TLR7. However, the importance of TLR7 processing in the development of anti-viral immune responses is not known. Here we report that asparagine endopeptidase (AEP) deficient mice are unable to generate a strong anti-IAV response, as demonstrated by reduced inflammation, cross presentation of cell-associated antigens and priming of CD8(+) T cells following TLR7-dependent pulmonary infection induced by IAV. Moreover, AEP deficient lung epithelial- or myeloid-cells exhibit impaired TLR7 signaling due to defective processing of this receptor. Indeed, TLR7 requires a proteolytic cleavage by AEP to generate a C-terminal fragment competent for signaling. Thus, AEP activity is critical for TLR7 processing, opening new possibilities for the treatment of influenza and TLR7-dependent inflammatory diseases.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Endopeptidasas/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Glicoproteínas de Membrana/inmunología , Infecciones por Orthomyxoviridae/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 7/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Endopeptidasas/genética , Endopeptidasas/metabolismo , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/metabolismo , Transducción de Señal/genética , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo
11.
Am J Respir Crit Care Med ; 187(2): 170-9, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23220915

RESUMEN

RATIONALE: Cystic fibrosis transmembrane conductance regulator (CFTR) protein is a chloride channel regulating fluid homeostasis at epithelial surfaces. Its loss of function induces hypohydration, mucus accumulation, and bacterial infections in CF and potentially other lung chronic diseases. OBJECTIVES: To test whether neutrophil elastase (NE) and neutrophil-mediated inflammation negatively impact CFTR structure and function, in vitro and in vivo. METHODS: Using an adenovirus-CFTR overexpression approach, we showed that NE degrades wild-type (WT)- and ΔF508-CFTR in vitro and WT-CFTR in mice through a new pathway involving the activation of intracellular calpains. MEASUREMENTS AND MAIN RESULTS: CFTR degradation triggered a loss of function, as measured in vitro by channel patch-clamp and in vivo by nasal potential recording in mice. Importantly, this mechanism was also shown to be operative in a Pseudomonas aeruginosa lung infection murine model, and was NE-dependent, because CFTR integrity was significantly protected in NE(-/-) mice compared with WT mice. CONCLUSIONS: These data provide a new mechanism and show for the first time a link between NE-calpains activation and CFTR loss of function in bacterial lung infections relevant to CF and to other chronic inflammatory lung conditions.


Asunto(s)
Calpaína/fisiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Elastasa de Leucocito/fisiología , Animales , Calpaína/metabolismo , Canales de Cloruro/fisiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Epitelio/fisiología , Humanos , Elastasa de Leucocito/metabolismo , Pulmón/metabolismo , Pulmón/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp , Neumonía Bacteriana/etiología , Neumonía Bacteriana/fisiopatología , Infecciones por Pseudomonas/etiología , Infecciones por Pseudomonas/fisiopatología
12.
BMC Pulm Med ; 14: 100, 2014 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-24909229

RESUMEN

BACKGROUND: The aim was to measure flagellin concentrations in the expectorations of CF patients and to examine whether there are correlations with the level of respiratory insufficiency and inflammation. METHODS: Sputum samples from 31 adult patients chronically colonized with P. aeruginosa were collected and analysed for their content of flagellin and IL-8. Clinical data were extracted from patient files. RESULTS: Regardless of whether patients are colonized with mucoid strains or not, they carry clones of P. aeruginosa that express flagellin. While flagellin was present in airways of all of our CF patients, it is difficult to ascertain its contribution to inflammation (IL-8) and lung function deterioration. CONCLUSIONS: This is the first demonstration that flagellin is present in the sputum of patients. Thus, attempts to down regulate inflammation by the use of TLR5 (flagellin receptor) antagonists remain a possibility. However, this result needs to be extended to a larger number of patients to validate it for future research on this subject.


Asunto(s)
Fibrosis Quística/microbiología , Flagelina/análisis , Infecciones por Pseudomonas/diagnóstico , Pseudomonas aeruginosa/aislamiento & purificación , Insuficiencia Respiratoria/diagnóstico , Esputo/metabolismo , Adulto , Biomarcadores/análisis , Fibrosis Quística/diagnóstico , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Interleucina-8/análisis , Masculino , Infecciones por Pseudomonas/epidemiología , Muestreo , Índice de Severidad de la Enfermedad , Esputo/microbiología
13.
PLoS Pathog ; 7(9): e1002238, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21909279

RESUMEN

Lectins and adhesins are involved in bacterial adhesion to host tissues and mucus during early steps of infection. We report the characterization of BC2L-C, a soluble lectin from the opportunistic pathogen Burkholderia cenocepacia, which has two distinct domains with unique specificities and biological activities. The N-terminal domain is a novel TNF-α-like fucose-binding lectin, while the C-terminal part is similar to a superfamily of calcium-dependent bacterial lectins. The C-terminal domain displays specificity for mannose and l-glycero-d-manno-heptose. BC2L-C is therefore a superlectin that binds independently to mannose/heptose glycoconjugates and fucosylated human histo-blood group epitopes. The apo form of the C-terminal domain crystallized as a dimer, and calcium and mannose could be docked in the binding site. The whole lectin is hexameric and the overall structure, determined by electron microscopy and small angle X-ray scattering, reveals a flexible arrangement of three mannose/heptose-specific dimers flanked by two fucose-specific TNF-α-like trimers. We propose that BC2L-C binds to the bacterial surface in a mannose/heptose-dependent manner via the C-terminal domain. The TNF-α-like domain triggers IL-8 production in cultured airway epithelial cells in a carbohydrate-independent manner, and is therefore proposed to play a role in the dysregulated proinflammatory response observed in B. cenocepacia lung infections. The unique architecture of this newly recognized superlectin correlates with multiple functions including bacterial cell cross-linking, adhesion to human epithelia, and stimulation of inflammation.


Asunto(s)
Mediadores de Inflamación/fisiología , Lectinas/fisiología , Secuencia de Aminoácidos , Burkholderia cenocepacia , Cristalografía por Rayos X , Fucosa/metabolismo , Humanos , Interleucina-8 , Lectinas/química , Lectinas/metabolismo , Lectinas de Unión a Manosa/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína/fisiología , Mucosa Respiratoria/citología , Mucosa Respiratoria/metabolismo , Alineación de Secuencia , Factor de Necrosis Tumoral alfa/metabolismo
14.
Am J Respir Cell Mol Biol ; 47(2): 149-57, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22383584

RESUMEN

Mucins, the main glycoproteins present within mucus, modulate the rheologic properties of airways and participate in lung defense. They are thought to be able to trap and eliminate microorganisms from the lung. Among the mucins secreted in the lung, MUC5AC is the most prominent factor secreted by surface epithelial cells. Although much is known about the signaling pathways involved in the regulation of MUC5AC by host factors such as cytokines or proteases, less is known about the pathways triggered by microorganisms and, specifically, by influenza A virus (IAV). We therefore set up experiments to dissect the molecular mechanisms responsible for the potential modulation of MUC5AC by IAV. Using epithelial cells, C57/Bl6 mice, and IAV strains, we measured MUC5AC expression at the RNA and protein levels, specificity protein 1 (Sp1) activation, and protease activity. Intermediate molecular partners were confirmed using pharmacological inhibitors, blocking antibodies, and small interfering (si)RNAs. We showed in vitro and in vivo that IAV up-regulates epithelial cell-derived MUC5AC and Muc5ac expression in mice, both at transcriptional (through the induction of Sp1) and translational levels. In addition, we determined that this induction was dependent on a protease-epithelial growth factor receptor-extracellular regulated kinase-Sp1 signaling cascade, involving in particular the human airway trypsin. Our data point to MUC5AC as a potential modulatory mechanism by which the lung epithelia respond to IAV infection, and we dissect, for the first time to the best of our knowledge, the molecular partners involved. Future experiments using MUC5AC-targeted strategies should help further unravel the pathophysiological consequences of IAV-induced MUC5AC expression for lung homeostasis.


Asunto(s)
Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Virus de la Influenza A/metabolismo , Pulmón/metabolismo , Mucina 5AC/biosíntesis , Péptido Hidrolasas/genética , Factor de Transcripción Sp1/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM17 , Animales , Línea Celular Tumoral , Células Epiteliales/metabolismo , Células Epiteliales/virología , Receptores ErbB/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Humanos , Gripe Humana/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mucina 5AC/genética , Mucina 5AC/metabolismo , Péptido Hidrolasas/metabolismo , Transducción de Señal , Factor de Transcripción Sp1/genética , Tripsina/genética , Tripsina/metabolismo , Regulación hacia Arriba , Replicación Viral/genética
15.
Gastroenterology ; 140(4): 1272-82, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21199654

RESUMEN

BACKGROUND & AIMS: Colonic tissues of patients with inflammatory bowel disease have been reported to have increased proteolytic activity, but no studies have clearly addressed the role of the balance between proteases and antiproteases in the pathogenesis of colitis. We investigated the role of Elafin, a serine protease inhibitor expressed by skin and mucosal surfaces in human inflammatory conditions, and the proteases neutrophil elastase (NE) and proteinase-3 (PR-3) in mice with colitis. METHODS: We studied mice with heterozygous disruptions in NE and PR-3, mice that express human elafin (an inhibitor of NE and PR-3), and naïve mice that received intracolonic adenoviral vectors that express elafin. Trinitrobenzene sulfonic acid (TNBS) or dextran sodium sulphate (DSS) was used to induce colitis. Protease, cytokine levels, and NF-κB activity were measured in colons of mice. Caco-2 and HT29 cells were studied in assays for cytokine expression, permeability, and NF-κB activity. RESULTS: Elafin expression or delivery re-equilibrated the proteolytic balance in inflamed colons of mice. In mice given TNBS or DSS, transgenic expression of elafin or disruption of NE and PR-3 protected against the development of colitis. Similarly, adenoviral delivery of Elafin significantly inhibited inflammatory parameters. Elafin modulated a variety of inflammatory mediators in vitro and in vivo and strengthened intestinal epithelial barrier functions. CONCLUSIONS: The protease inhibitor Elafin prevents intestinal inflammation in mouse models of colitis and might be developed as a therapeutic agent for inflammatory bowel disease.


Asunto(s)
Colitis , Elafina/genética , Terapia Genética/métodos , Elastasa de Leucocito/metabolismo , Inhibidores de Proteasas/metabolismo , Adenoviridae/genética , Animales , Células CACO-2 , Quimiocinas/metabolismo , Colitis/genética , Colitis/metabolismo , Colitis/terapia , Citocinas/metabolismo , Elafina/metabolismo , Expresión Génica/fisiología , Células HT29 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Mieloblastina/metabolismo , FN-kappa B/metabolismo , Neutrófilos/enzimología , Neutrófilos/inmunología , Inhibidores de Serina Proteinasa/metabolismo
16.
Eur Respir J ; 40(3): 714-23, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22441738

RESUMEN

Eicosanoids are metabolites of arachidonic acid produced by cyclooxygenases (COXs) or lipoxygenases (LOXs). They mediate inflammation and mucus secretion in chronic pulmonary inflammatory diseases. The gel-forming mucin MUC5AC is over-expressed in the airways of patients with these diseases. MUC5AC expression is mediated by an extracellular signal-regulated kinase (ERK)/Sp1 dependent mechanism. Our aim was to study the role of eicosanoids and their signalling pathways in MUC5AC expression. Inhibitors of 12-LOX, but not those of COX, 5-LOX or 15-LOX, reduce MUC5AC expression induced by phorbol myristate acetate (PMA) in the bronchial epithelial cell line NCI-H292. These inhibitors also abrogate the production of whole mucus by cell monolayers. Two forms of 12-LOX (R and S) exist in mammals. Using siRNAs we show that 12R-LOX but not 12S-LOX is involved in MUC5AC expression induced by PMA, lipopolysaccharide or transforming growth factor-α. 12R-LOX also participates in MUC2 and MUC5B expression, although to a lesser extent than for MUC5AC. Contrarily, 12R-LOX silencing does not modify interleukin-8 production. 12-LOX inhibitors reduce ERK activation and Sp1 translocation induced by PMA. Moreover, the 12R-LOX product 12(R)-hydroxyeicosatetraenoic acid, induces MUC5AC expression, ERK activation and Sp1 translocation. 12R-LOX is involved in MUC5AC expression. This occurs via ERK- and Sp1-signalling pathways.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Mucina 5AC/biosíntesis , Mucosa Respiratoria/metabolismo , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/metabolismo , Araquidonato 12-Lipooxigenasa/genética , Carcinógenos/farmacología , Línea Celular , Inhibidores de la Ciclooxigenasa/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Silenciador del Gen , Humanos , Interleucina-8/biosíntesis , Lipopolisacáridos/farmacología , Inhibidores de la Lipooxigenasa/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mucina 2/biosíntesis , Mucina 5B/biosíntesis , Moco/metabolismo , Transporte de Proteínas , Mucosa Respiratoria/efectos de los fármacos , Factor de Transcripción Sp1/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Factor de Crecimiento Transformador alfa/farmacología
17.
J Immunol ; 184(4): 2038-47, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20083674

RESUMEN

Early immune response to the largely used Mycobacterium bovis bacillus Calmette-Guérin (BCG) intradermal vaccine remains ill defined. Three days after BCG inoculation into the mouse ear, in addition to neutrophils infiltrating skin, we observed CD11b(+)Ly-6C(int)Ly-6G(-) myeloid cells. Neutrophil depletion markedly enhanced their recruitment. These cells differed from inflammatory monocytes and required MyD88-dependent BCG-specific signals to invade skin, whereas neutrophil influx was MyD88 independent. Upon BCG phagocytosis, CD11b(+)Ly-6C(int)Ly-6G(-) cells produced NO, which required the IL-1 receptor. Despite NO production, they were unable to kill BCG or the nonpathogenic Mycobacterium smegmatis. However, they markedly impaired T cell priming in the draining lymph node. Their elimination by all-trans retinoid acid treatment increased the number of IFN-gamma-producing CD4 T cells. Thus, BCG vaccination recruits innate myeloid-derived suppressor cells, akin to mouse tumor-infiltrating cells. These propathogenic cells dampen the early T cell response and might facilitate BCG persistence.


Asunto(s)
Vacuna BCG/administración & dosificación , Vacuna BCG/inmunología , Movimiento Celular/inmunología , Activación de Linfocitos/inmunología , Células Mieloides/inmunología , Óxido Nítrico/biosíntesis , Receptores de Interleucina-1/fisiología , Subgrupos de Linfocitos T/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Femenino , Inmunidad Innata , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/citología , Neutrófilos/inmunología , Neutrófilos/patología , Óxido Nítrico/fisiología , Receptores de Interleucina-1/antagonistas & inhibidores , Subgrupos de Linfocitos T/patología
18.
J Infect Dis ; 203(10): 1369-77, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21502078

RESUMEN

BACKGROUND: The role of toxins secreted by the type II secretion system (T2SS) of Pseudomonas aeruginosa during lung infection has been uncertain despite decades of research. METHODS: Using a model of pneumonia in Toll-like receptor (TLR) 2,4(-/-) mice, we reexamined the role of the T2SS system. Flagellin-deficient mutants of P. aeruginosa, with mutations in the T2SS and/or T3SS, were used to infect mice. Mice were followed up for survival, with some killed at different intervals to study bacterial clearance, inflammatory responses, and lung pathology. RESULTS: Strains carrying either secretion system were lethal for mice. Double mutants were avirulent. The T3SS(+) strains killed mice within a day, and the T2SS(+) strains killed them later. Mice infected with a strain that had only the T2SS were unable to eradicate the organism from the lungs, whereas those infected with a T2SS-T3SS double deletion were able to clear this mutant. Death caused by the T2SS(+) strain was accompanied by a >50-fold increase in bacterial counts and higher numbers of viable intracellular bacteria. CONCLUSIONS: The T2SS of P. aeruginosa may play a role in death from pneumonia, but its action is delayed. These data suggest that antitoxin strategies against this organism will require measures against the toxins secreted by both T2SS and T3SS.


Asunto(s)
Toxinas Bacterianas/metabolismo , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/mortalidad , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/mortalidad , Pseudomonas aeruginosa/genética , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/microbiología , Flagelina/genética , Flagelina/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Inmunidad Innata , Inmunocompetencia , Pulmón/patología , Ratones , Ratones Noqueados , Mutación , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidad , Secretina/fisiología , Factores de Tiempo , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
19.
J Immunol ; 183(1): 670-6, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19535624

RESUMEN

Burkholderia cenocepacia is an opportunistic pathogen of major concern for cystic fibrosis patients as well as immunocompromised cancer patients and transplant recipients. The mechanisms by which B. cenocepacia triggers a rapid health deterioration of the susceptible host have yet to be characterized. TLR and their key signaling intermediate MyD88 play a central role in the detection of microbial molecular patterns and in the initiation of an effective immune response. We performed a study to better understand the role of TLR-MyD88 signaling in B. cenocepacia-induced pathogenesis in the immunocompromised host, using an experimental murine model. The time-course of several dynamic parameters, including animal survival, bacterial load, and secretion of critical inflammatory mediators, was compared in infected and immunosuppressed wild-type and MyD88(-/-) mice. Notably, when compared with wild-type mice, infected MyD88(-/-) animals displayed significantly reduced levels of inflammatory mediators (including KC, TNF-alpha, IL-6, MIP-2, and G-CSF) in blood and lung airspaces. Moreover, despite a higher transient bacterial load in the lungs, immunosuppressed mice deficient in MyD88 had an unexpected survival advantage. Finally, we showed that this B. cenocepacia-induced life-threatening infection of wild-type mice involved the proinflammatory cytokine TNF-alpha and could be prevented by corticosteroids. Altogether, our findings demonstrate that a MyD88-dependent pathway can critically contribute to a detrimental host inflammatory response that leads to fatal pneumonia.


Asunto(s)
Infecciones por Burkholderia/prevención & control , Burkholderia cepacia , Huésped Inmunocomprometido/genética , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Infecciones Oportunistas/prevención & control , Neumonía Bacteriana/prevención & control , Animales , Infecciones por Burkholderia/inmunología , Infecciones por Burkholderia/mortalidad , Burkholderia cepacia/crecimiento & desarrollo , Burkholderia cepacia/inmunología , Burkholderia cepacia/patogenicidad , Huésped Inmunocomprometido/inmunología , Inmunosupresores/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/fisiología , Infecciones Oportunistas/inmunología , Infecciones Oportunistas/mortalidad , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/mortalidad , Análisis de Supervivencia , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/fisiología
20.
Respir Res ; 11: 49, 2010 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-20429932

RESUMEN

BACKGROUND: Lungs of cystic fibrosis (CF) patients are chronically infected with Pseudomonas aeruginosa. Increased airway constriction has been reported in CF patients but underplaying mechanisms have not been elucidated. AIM: To examine the effect of P. aeruginosa LPS on airway constriction in CF mice and the implication in this process of cytosolic phospholipase A2alpha (cPLA2alpha), an enzyme involved in arachidonic acid (AA) release. METHODS: Mice were instilled intra-nasally with LPS. Airway constriction was assessed using barometric plethysmograph. MIP-2, prostaglandin E2 (PGE2), leukotrienes and AA concentrations were measured in BALF using standard kits and gas chromatography. RESULTS: LPS induced enhanced airway constriction and AA release in BALF of CF compared to littermate mice. This was accompanied by increased levels of PGE2, but not those of leukotrienes. However, airway neutrophil influx and MIP-2 production remained similar in both mouse strains. The cPLA2alpha inhibitor arachidonyl trifluoro-methyl-ketone (ATK), but not aspirin which inhibit PGE2 synthesis, reduced LPS-induced airway constriction. LPS induced lower airway constriction and PGE2 production in cPLA2alpha -/- mice compared to corresponding littermates. Neither aspirin nor ATK interfered with LPS-induced airway neutrophil influx or MIP-2 production. CONCLUSIONS: CF mice develop enhanced airway constriction through a cPLA2alpha-dependent mechanism. Airway inflammation is dissociated from airway constriction in this model. cPLA2alpha may represent a suitable target for therapeutic intervention in CF. Attenuation of airway constriction by cPLA2alpha inhibitors may help to ameliorate the clinical status of CF patients.


Asunto(s)
Broncoconstricción/efectos de los fármacos , Fibrosis Quística/enzimología , Fosfolipasas A2 Grupo IV/metabolismo , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Pseudomonas aeruginosa , Administración Intranasal , Animales , Ácido Araquidónico/metabolismo , Ácidos Araquidónicos/farmacología , Aspirina/farmacología , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Quimiocina CXCL2/metabolismo , Inhibidores de la Ciclooxigenasa/farmacología , Fibrosis Quística/inmunología , Fibrosis Quística/fisiopatología , Dinoprostona/metabolismo , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Fosfolipasas A2 Grupo IV/antagonistas & inhibidores , Fosfolipasas A2 Grupo IV/deficiencia , Fosfolipasas A2 Grupo IV/genética , Leucotrienos/metabolismo , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/aislamiento & purificación , Pulmón/enzimología , Pulmón/inmunología , Pulmón/fisiopatología , Ratones , Ratones Endogámicos CFTR , Ratones Noqueados , Infiltración Neutrófila/efectos de los fármacos , Neumonía/enzimología , Neumonía/inmunología , Neumonía/fisiopatología , Pseudomonas aeruginosa/química , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA