Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 114(8): E1519-E1527, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28167784

RESUMEN

Bacterial pathogens coordinate virulence using two-component regulatory systems (TCS). The Bordetella virulence gene (BvgAS) phosphorelay-type TCS controls expression of all known protein virulence factor-encoding genes and is considered the "master virulence regulator" in Bordetella pertussis, the causal agent of pertussis, and related organisms, including the broad host range pathogen Bordetella bronchiseptica We recently discovered an additional sensor kinase, PlrS [for persistence in the lower respiratory tract (LRT) sensor], which is required for B. bronchiseptica persistence in the LRT. Here, we show that PlrS is required for BvgAS to become and remain fully active in mouse lungs but not the nasal cavity, demonstrating that PlrS coordinates virulence specifically in the LRT. PlrS is required for LRT persistence even when BvgAS is rendered constitutively active, suggesting the presence of BvgAS-independent, PlrS-dependent virulence factors that are critical for bacterial survival in the LRT. We show that PlrS is also required for persistence of the human pathogen B. pertussis in the murine LRT and we provide evidence that PlrS most likely functions via the putative cognate response regulator PlrR. These data support a model in which PlrS senses conditions present in the LRT and activates PlrR, which controls expression of genes required for the maintenance of BvgAS activity and for essential BvgAS-independent functions. In addition to providing a major advance in our understanding of virulence regulation in Bordetella, which has served as a paradigm for several decades, these results indicate the existence of previously unknown virulence factors that may serve as new vaccine components and therapeutic or diagnostic targets.


Asunto(s)
Proteínas Bacterianas/genética , Bordetella bronchiseptica/genética , Bordetella pertussis/patogenicidad , Regulación Bacteriana de la Expresión Génica , Sistema Respiratorio/microbiología , Factores de Virulencia/genética , Animales , Proteínas Bacterianas/metabolismo , Bordetella pertussis/genética , Línea Celular , Femenino , Ratones , Ratones Endogámicos BALB C , Ratas , Virulencia , Factores de Virulencia/metabolismo
2.
Pathog Dis ; 73(8): ftv079, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26416077

RESUMEN

Pertussis, or whooping cough, is a highly contagious respiratory disease that is caused by the Gram-negative bacterium Bordetella pertussis, which is transmitted exclusively from human to human. While vaccination against B. pertussis has been successful, replacement of the whole cell vaccine with an acellular component vaccine has correlated with reemergence of the disease, especially in adolescents and infants. Based on their presumed importance in mediating adherence to host tissues, filamentous hemagglutinin (FHA) and fimbria (FIM) were selected as components of most acellular pertussis vaccines. In this review, we describe the biogenesis of FHA and FIM, recent data that show that these factors do, in fact, play critical roles in adherence to respiratory epithelium, and evidence that they also contribute to persistence in the lower respiratory tract by modulating the host immune response. We also discuss shortcomings of whole cell and acellular pertussis vaccines and the possibility that FHA and FIM could serve as effective protective antigens in next-generation vaccines.


Asunto(s)
Adhesinas Bacterianas/inmunología , Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana , Bordetella pertussis/inmunología , Bordetella pertussis/fisiología , Fimbrias Bacterianas/inmunología , Fimbrias Bacterianas/metabolismo , Factores de Virulencia de Bordetella/inmunología , Factores de Virulencia de Bordetella/metabolismo , Humanos , Vacuna contra la Tos Ferina/inmunología
3.
mBio ; 6(4)2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26286694

RESUMEN

UNLABELLED: Bordetella filamentous hemagglutinin (FHA), a primary component of acellular pertussis vaccines, contributes to virulence, but how it functions mechanistically is unclear. FHA is first synthesized as an ~370-kDa preproprotein called FhaB. Removal of an N-terminal signal peptide and a large C-terminal prodomain (PD) during secretion results in "mature" ~250-kDa FHA, which has been assumed to be the biologically active form of the protein. Deletion of two C-terminal subdomains of FhaB did not affect production of functional FHA, and the mutant strains were indistinguishable from wild-type bacteria for their ability to adhere to the lower respiratory tract and to suppress inflammation in the lungs of mice. However, the mutant strains, which produced altered FhaB molecules, were eliminated from the lower respiratory tract much faster than wild-type B. bronchiseptica, suggesting a defect in resistance to early immune-mediated clearance. Our results revealed, unexpectedly, that full-length FhaB plays a critical role in B. bronchiseptica persistence in the lower respiratory tract. IMPORTANCE: The Bordetella filamentous hemagglutinin (FHA) is a primary component of the acellular pertussis vaccine and an important virulence factor. FHA is initially produced as a large protein that is processed during secretion to the bacterial surface. As with most processed proteins, the mature form of FHA has been assumed to be the functional form of the protein. However, our results indicate that the full-length form plays an essential role in virulence in vivo. Furthermore, we have found that FHA contains intramolecular regulators of processing and that this control of processing is integral to its virulence activities. This report highlights the advantage of studying protein maturation and function simultaneously, as a role for the full-length form of FHA was evident only from in vivo infection studies and not from in vitro studies on the production or maturation of FHA or even from in vitro virulence-associated activity assays.


Asunto(s)
Adhesinas Bacterianas/química , Adhesinas Bacterianas/metabolismo , Bordetella bronchiseptica/metabolismo , Bordetella bronchiseptica/patogenicidad , Mucosa Respiratoria/microbiología , Factores de Virulencia de Bordetella/química , Factores de Virulencia de Bordetella/metabolismo , Adhesinas Bacterianas/genética , Animales , Adhesión Bacteriana , Bordetella bronchiseptica/genética , Bordetella bronchiseptica/inmunología , Vacunas contra Difteria, Tétanos y Tos Ferina Acelular/química , Femenino , Pulmón/microbiología , Ratones , Mutación , Estructura Terciaria de Proteína , Alineación de Secuencia , Virulencia/genética , Factores de Virulencia de Bordetella/genética
4.
Am J Physiol Lung Cell Mol Physiol ; 309(2): L158-67, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26001778

RESUMEN

Suppression of type 17 immunity by type I interferon (IFN) during influenza A infection has been shown to enhance susceptibility to secondary bacterial pneumonia. Although this mechanism has been described in coinfection with gram-positive bacteria, it is unclear whether similar mechanisms may impair lung defense against gram-negative infections. Furthermore, precise delineation of the duration of type I IFN-associated susceptibility to bacterial infection remains underexplored. Therefore, we investigated the effects of preceding influenza A virus infection on subsequent challenge with the gram-negative bacteria Escherichia coli or Pseudomonas aeruginosa and the temporal association between IFN expression with susceptibility to Staphylococcus aureus challenge in a mouse model of influenza and bacterial coinfection. Here we demonstrate that preceding influenza A virus led to increased lung E. coli and P. aeruginosa bacterial burden, which was associated with suppression of type 17 immunity and attenuation of antimicrobial peptide expression. Enhanced susceptibility to S. aureus coinfection ceased at day 14 of influenza infection, when influenza-associated type I IFN levels had returned to baseline levels, further suggesting a key role for type I IFN in coinfection pathogenesis. These findings further implicate type I IFN-associated suppression of type 17 immunity and antimicrobial peptide production as a conserved mechanism for enhanced susceptibility to both gram-positive and gram-negative bacterial coinfection during influenza infection.


Asunto(s)
Infecciones por Escherichia coli/microbiología , Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/microbiología , Neumonía Bacteriana/microbiología , Neumonía/microbiología , Receptor de Interferón alfa y beta/fisiología , Infecciones Estafilocócicas/microbiología , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Coinfección/inmunología , Coinfección/microbiología , Coinfección/virología , Susceptibilidad a Enfermedades , Escherichia coli/inmunología , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/virología , Virus de la Influenza A/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Neumonía/inmunología , Neumonía/virología , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/virología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/virología , Staphylococcus aureus/inmunología , Staphylococcus aureus/patogenicidad
5.
mBio ; 6(3): e00500-15, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-26015497

RESUMEN

UNLABELLED: Bordetella fimbriae (FIM) are generally considered to function as adhesins despite a lack of experimental evidence supporting this conclusion for Bordetella pertussis and evidence against a requirement for FIM in adherence of Bordetella bronchiseptica to mammalian cell lines. Using B. bronchiseptica and mice, we developed an in vivo adherence assay that revealed that FIM do function as critically important adhesins in the lower respiratory tract. In the first few days postinoculation, FIM-deficient B. bronchiseptica induced a more robust inflammatory response than wild-type bacteria did, suggesting that FIM, like filamentous hemagglutinin (FHA), allow B. bronchiseptica to suppress the innate immune response to infection. Localization analyses indicated that FIM are required for efficient attachment to airway epithelium, as bacteria lacking FIM localized to alveoli. FHA-deficient bacteria, in contrast, localized to airways. Bacteria unable to produce both FIM and FHA localized to alveoli and caused increased inflammation and histopathology identical to that caused by FIM-deficient bacteria, demonstrating that lack of FIM is epistatic to lack of FHA. Coinoculation experiments provided evidence that wild-type B. bronchiseptica suppresses inflammation locally within the respiratory tract and that both FHA and FIM are required for defense against clearance by the innate immune system. Altogether, our data suggest that FIM-mediated adherence to airway epithelium is a critical first step in Bordetella infection that allows FHA-dependent interactions to mediate tight adherence, suppression of inflammation, and resistance to inflammatory cell-mediated clearance. Our results suggest that mucosal antibodies capable of blocking FIM-mediated interactions could prevent bacterial colonization of the lower respiratory tract. IMPORTANCE: Although fimbriae (FIM) have been shown to be important mediators of adherence for many bacterial pathogens, there is surprisingly little experimental evidence supporting this role for Bordetella fimbria. Our results provide the first demonstration that Bordetella FIM function as adhesins in vivo, specifically to airway epithelium. Furthermore, our results suggest that FIM mediate initial interactions with airway epithelial cells that are followed by tight filamentous hemagglutinin (FHA)-mediated binding and that together, FIM and FHA allow Bordetella to suppress inflammation, leading to prolonged colonization. Given the shortcoming of the current acellular component pertussis (aP) vaccine in preventing colonization, these findings suggest that generation of antibodies capable of blocking FIM-mediated adherence could potentially prevent Bordetella colonization.


Asunto(s)
Adhesinas Bacterianas/fisiología , Adhesión Bacteriana , Infecciones por Bordetella/inmunología , Infecciones por Bordetella/microbiología , Bordetella bronchiseptica/fisiología , Fimbrias Bacterianas/fisiología , Adhesinas Bacterianas/inmunología , Animales , Bordetella bronchiseptica/inmunología , Bordetella bronchiseptica/patogenicidad , Línea Celular , Células Epiteliales/microbiología , Fimbrias Bacterianas/inmunología , Inmunidad Innata , Pulmón/inmunología , Pulmón/microbiología , Pulmón/ultraestructura , Ratones , Ratones Endogámicos BALB C , Mutación , Tráquea/microbiología , Factores de Virulencia de Bordetella/inmunología
6.
Respir Res ; 16: 10, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25651926

RESUMEN

BACKGROUND: Influenza is a common respiratory virus and Staphylococcus aureus frequently causes secondary pneumonia during influenza infection, leading to increased morbidity and mortality. Influenza has been found to attenuate subsequent Type 17 immunity, enhancing susceptibility to secondary bacterial infections. IL-27 is known to inhibit Type 17 immunity, suggesting a potential critical role for IL-27 in viral and bacterial co-infection. METHODS: A murine model of influenza and Staphylococcus aureus infection was used to mimic human viral, bacterial co-infection. C57BL/6 wild-type, IL-27 receptor α knock-out, and IL-10 knock-out mice were infected with Influenza H1N1 (A/PR/8/34) or vehicle for 6 days followed by challenge with Staphylococcus aureus or vehicle for 24 hours. Lung inflammation, bacterial burden, gene expression, and cytokine production were determined. RESULTS: IL-27 receptor α knock-out mice challenged with influenza A had increased morbidity compared to controls, but no change in viral burden. IL-27 receptor α knock-out mice infected with influenza displayed significantly decreased IL-10 production compared to wild-type. IL-27 receptor α knock-out mice co-infected with influenza and S. aureus had improved bacterial clearance compared to wild-type controls. Importantly, there were significantly increased Type 17 responses and decreased IL-10 production in IL-27 receptor α knock-out mice. Dual infected IL-10-/- mice had significantly less bacterial burden compared to dual infected WT mice. CONCLUSIONS: These data reveal that IL-27 regulates enhanced susceptibility to S. aureus pneumonia following influenza infection, potentially through the induction of IL-10 and suppression of IL-17.


Asunto(s)
Coinfección , Subtipo H1N1 del Virus de la Influenza A/inmunología , Interleucinas/inmunología , Pulmón/inmunología , Infecciones por Orthomyxoviridae/inmunología , Neumonía Estafilocócica/inmunología , Staphylococcus aureus/inmunología , Animales , Carga Bacteriana , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno , Inmunidad Celular , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Interleucina-10/deficiencia , Interleucina-10/genética , Interleucina-17/inmunología , Pulmón/microbiología , Pulmón/virología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/virología , Neumonía Estafilocócica/genética , Neumonía Estafilocócica/microbiología , Receptores de Citocinas/deficiencia , Receptores de Citocinas/genética , Receptores de Interleucina , Staphylococcus aureus/patogenicidad , Células Th17/inmunología , Células Th17/microbiología , Células Th17/virología , Factores de Tiempo , Carga Viral
7.
Nat Rev Microbiol ; 12(4): 274-88, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24608338

RESUMEN

Pertussis, also known as whooping cough, has recently re-emerged as a major public health threat despite high levels of vaccination against the aetiological agent Bordetella pertussis. In this Review, we describe the pathogenesis of this disease, with a focus on recent mechanistic insights into B. pertussis virulence-factor function. We also discuss the changing epidemiology of pertussis and the challenges facing vaccine development. Despite decades of research, many aspects of B. pertussis physiology and pathogenesis remain poorly understood. We highlight knowledge gaps that must be addressed to develop improved vaccines and therapeutic strategies.


Asunto(s)
Bordetella pertussis/patogenicidad , Enfermedades Transmisibles Emergentes/microbiología , Tos Ferina/microbiología , Investigación Biomédica/tendencias , Enfermedades Transmisibles Emergentes/epidemiología , Humanos , Microbiología/tendencias , Vacuna contra la Tos Ferina/inmunología , Vacuna contra la Tos Ferina/aislamiento & purificación , Virulencia , Factores de Virulencia/metabolismo , Tos Ferina/epidemiología
8.
J Infect Dis ; 209(6): 865-75, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24072844

RESUMEN

Influenza A represents a significant cause of morbidity and mortality worldwide. Bacterial complications of influenza A confer the greatest risk to patients. TH17 pathway inhibition has been implicated as a mechanism by which influenza A alters bacterial host defense. Here we show that preceding influenza causes persistent Staphylococcus aureus infection and suppression of TH17 pathway activation in mice. Influenza does not inhibit S. aureus binding and uptake by phagocytic cells but instead attenuates S. aureus induced TH17 related antimicrobial peptides necessary for bacterial clearance in the lung. Importantly, exogenous lipocalin 2 rescued viral exacerbation of S. aureus infection and decreased free iron levels in the bronchoalveolar lavage from mice coinfected with S. aureus and influenza. These findings indicate a novel mechanism by which influenza A inhibits TH17 immunity and increases susceptibility to secondary bacterial pneumonia. Identification of new mechanisms in the pathogenesis of bacterial pneumonia could lead to future therapeutic targets.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/microbiología , Neumonía Estafilocócica/microbiología , Staphylococcus aureus/inmunología , Análisis de Varianza , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/microbiología , Líquido del Lavado Bronquioalveolar/virología , Coinfección/microbiología , Coinfección/virología , Interacciones Huésped-Patógeno/inmunología , Virus de la Influenza A/patogenicidad , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Neumonía Estafilocócica/inmunología , Neumonía Estafilocócica/virología , Staphylococcus aureus/patogenicidad , Células Th17
9.
Mol Microbiol ; 90(4): 716-33, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24007341

RESUMEN

Bordetella species cause respiratory infections in mammals. Their master regulatory system BvgAS controls expression of at least three distinct phenotypic phases in response to environmental cues. The Bvg⁺ phase is necessary and sufficient for respiratory infection while the Bvg⁻ phase is required for survival ex vivo. We obtained large colony variants (LCVs) from the lungs of mice infected with B. bronchiseptica strain RBX9, which contains an in-frame deletion mutation in fhaB, encoding filamentous haemagglutinin. RBX9 also yielded LCVs when switched from Bvg⁻ phase conditions to Bvg⁺ phase conditions in vitro. We determined that LCVs are composed of both Bvg⁺ and Bvg⁻ phase bacteria and that they result from defective bvgAS positive autoregulation. The LCV phenotype was linked to the presence of a divergent promoter 5' to bvgAS, suggesting a previously undescribed mechanism of transcriptional interference that, in this case, leads to feedback-based bistability (FBM). Our results also indicate that a small proportion of RBX9 bacteria modulates to the Bvg⁻ phase in vivo. In addition to providing insight into transcriptional interference and FBM, our data provide an example of an in-frame deletion mutation exerting a 'polar' effect on nearby genes.


Asunto(s)
Proteínas Bacterianas/metabolismo , Infecciones por Bordetella/microbiología , Bordetella bronchiseptica/genética , Regulación Bacteriana de la Expresión Génica , Pulmón/microbiología , Infecciones del Sistema Respiratorio/microbiología , Factores de Virulencia de Bordetella/genética , Animales , Proteínas Bacterianas/genética , Bordetella bronchiseptica/metabolismo , Bordetella bronchiseptica/patogenicidad , Modelos Animales de Enfermedad , Escherichia coli/enzimología , Escherichia coli/genética , Retroalimentación Fisiológica , Humanos , Ratones , Ratones Endogámicos BALB C , Fenotipo , Eliminación de Secuencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
10.
Am J Pathol ; 182(4): 1286-96, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23490254

RESUMEN

Influenza infection is widespread in the United States and the world. Despite low mortality rates due to infection, morbidity is common and little is known about the molecular events involved in recovery. Influenza infection results in persistent distal lung remodeling, and the mechanism(s) involved are poorly understood. Recently IL-22 has been found to mediate epithelial repair. We propose that IL-22 is critical for recovery of normal lung function and architecture after influenza infection. Wild-type and IL-22(-/-) mice were infected with influenza A PR8/34 H1N1 and were followed up for up to 21 days post infection. IL-22 receptor was localized to the airway epithelium in naive mice but was expressed at the sites of parenchymal lung remodeling induced by influenza infection. IL-22(-/-) mice displayed exacerbated lung injury compared with wild-type mice, which correlated with decreased lung function 21 days post infection. Epithelial metaplasia was observed in wild-type mice but was not evident in IL-22(-/-) animals that were characterized with an increased fibrotic phenotype. Gene expression analysis revealed aberrant expression of epithelial genes involved in repair processes, among changes in several other biological processes. These data indicate that IL-22 is required for normal lung repair after influenza infection. IL-22 represents a novel pathway involved in interstitial lung disease.


Asunto(s)
Epitelio/patología , Epitelio/virología , Interleucinas/metabolismo , Pulmón/patología , Pulmón/virología , Infecciones por Orthomyxoviridae/patología , Cicatrización de Heridas , Animales , Membrana Basal/metabolismo , Membrana Basal/patología , Colágeno/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Epiteliales/virología , Epitelio/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Interleucinas/deficiencia , Pulmón/fisiopatología , Metaplasia , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/fisiopatología , Infecciones por Orthomyxoviridae/virología , Receptores de Interleucina/metabolismo , Pruebas de Función Respiratoria , Transducción de Señal/genética , Interleucina-22
11.
Infect Immun ; 81(4): 1295-305, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23381998

RESUMEN

Bordetella pertussis and Bordetella bronchiseptica rely on the global two-component regulatory system BvgAS to control expression of distinct phenotypic phases. In the Bvg(-) phase, expression of vrg genes, including those required for motility in B. bronchiseptica, is activated and genes encoding virulence factors are not expressed. Conversely, in the Bvg(+) phase, genes encoding virulence factors are highly expressed while genes necessary for motility are repressed. Although several genetic analyses have demonstrated the importance of the Bvg(+) phase during respiratory infection, Bvg-regulated gene activation in B. bronchiseptica has not been investigated in vivo. To address this, we developed a plasmid, pGFLIP, that encodes a sensitive Flp recombinase-based fluorescent reporter system able to document gene activation both in vitro and in vivo. Using pGFLIP, we demonstrated that cyaA, considered to be a "late" Bvg(+) phase gene, is activated substantially earlier in B. bronchiseptica than B. pertussis following a switch from Bvg(-) to Bvg(+) phase conditions. We show that the altered activation of cyaA is not due to differences in the cyaA promoter or in the bvgAS alleles of B. bronchiseptica compared to B. pertussis, but appears to be species specific. Finally, we used pGFLIP to show that flaA remains repressed during infection, confirming that B. bronchiseptica does not modulate to the Bvg(-) phase in vivo.


Asunto(s)
Toxina de Adenilato Ciclasa/biosíntesis , Bordetella bronchiseptica/genética , Bordetella pertussis/genética , Regulación Bacteriana de la Expresión Génica , Factores de Virulencia/biosíntesis , Toxina de Adenilato Ciclasa/genética , Experimentación Animal , Animales , Bordetella bronchiseptica/patogenicidad , Bordetella pertussis/patogenicidad , Expresión Génica , Genes Reporteros , Genética Microbiana/métodos , Ratones , Ratones Endogámicos BALB C , Biología Molecular/métodos , Plásmidos , Recombinación Genética , Activación Transcripcional , Factores de Virulencia/genética
12.
PLoS One ; 7(4): e34638, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22514650

RESUMEN

The c-Jun N-terminal kinase (JNK) - 1 pathway has been implicated in the cellular response to stress in many tissues and models. JNK1 is known to play a role in a variety of signaling cascades, including those involved in lung disease pathogenesis. Recently, a role for JNK1 signaling in immune cell function has emerged. The goal of the present study was to determine the role of JNK1 in host defense against both bacterial and viral pneumonia, as well as the impact of JNK1 signaling on IL-17 mediated immunity. Wild type (WT) and JNK1 -/- mice were challenged with Escherichia coli, Staphylococcus aureus, or Influenza A. In addition, WT and JNK1 -/- mice and epithelial cells were stimulated with IL-17A. The impact of JNK1 deletion on pathogen clearance, inflammation, and histopathology was assessed. JNK1 was required for clearance of E. coli, inflammatory cell recruitment, and cytokine production. Interestingly, JNK1 deletion had only a small impact on the host response to S. aureus. JNK1 -/- mice had decreased Influenza A burden in viral pneumonia, yet displayed worsened morbidity. Finally, JNK1 was required for IL-17A mediated induction of inflammatory cytokines and antimicrobial peptides both in epithelial cells and the lung. These data identify JNK1 as an important signaling molecule in host defense and demonstrate a pathogen specific role in disease. Manipulation of the JNK1 pathway may represent a novel therapeutic target in pneumonia.


Asunto(s)
Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Neumonía/metabolismo , Animales , Células Cultivadas , Interleucina-17/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/microbiología , Ratones , Ratones Noqueados , Transducción de Señal/fisiología , Staphylococcus aureus/patogenicidad
13.
J Allergy Ther ; 2012(Suppl 1)2012 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-23560246

RESUMEN

Asthma represents a growing problem in the developing world, affecting millions of children and adults. Features of the disease are reversible airflow obstruction, airway hyperresponsiveness and airway inflammation leading to tissue damage and remodeling. Many studies have attempted to address whether inflammation and airway hyperresponsiveness are mechanistically linked. In this study, data are presented from several mouse models that illustrate that a clear link between these features of asthma remains elusive. The impact of altering inflammatory signaling (NF-κB or JNK1) on inflammation and airway hyperresponsiveness was examined. In addition, the effect of antigen sensitization and the route of antigen delivery were investigated. The data herein show that in many cases, inflammation and airway hyperresponsiveness do not directly correlate. In conclusion, the need for mechanistic studies in mouse models is highlighted to address the interplay between these components thought to be critical to asthma pathogenesis.

14.
J Immunol ; 186(3): 1666-1674, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21178015

RESUMEN

Staphylococcus aureus is a significant cause of hospital and community acquired pneumonia and causes secondary infection after influenza A. Recently, patients with hyper-IgE syndrome, who often present with S. aureus infections of the lung and skin, were found to have mutations in STAT3, required for Th17 immunity, suggesting a potential critical role for Th17 cells in S. aureus pneumonia. Indeed, IL-17R(-/-) and IL-22(-/-) mice displayed impaired bacterial clearance of S. aureus compared with that of wild-type mice. Mice challenged with influenza A PR/8/34 H1N1 and subsequently with S. aureus had increased inflammation and decreased clearance of both virus and bacteria. Coinfection resulted in greater type I and II IFN production in the lung compared with that with virus infection alone. Importantly, influenza A coinfection resulted in substantially decreased IL-17, IL-22, and IL-23 production after S. aureus infection. The decrease in S. aureus-induced IL-17, IL-22, and IL-23 was independent of type II IFN but required type I IFN production in influenza A-infected mice. Furthermore, overexpression of IL-23 in influenza A, S. aureus-coinfected mice rescued the induction of IL-17 and IL-22 and markedly improved bacterial clearance. These data indicate a novel mechanism by which influenza A-induced type I IFNs inhibit Th17 immunity and increase susceptibility to secondary bacterial pneumonia.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/inmunología , Interleucina-17/antagonistas & inhibidores , Interleucina-17/fisiología , Infecciones por Orthomyxoviridae/inmunología , Neumonía Bacteriana/inmunología , Infecciones Estafilocócicas/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Células Cultivadas , Predisposición Genética a la Enfermedad , Humanos , Interferón Tipo I/administración & dosificación , Interferón Tipo I/biosíntesis , Interleucina-17/deficiencia , Interleucina-23/antagonistas & inhibidores , Interleucinas/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/microbiología , Neumonía Bacteriana/genética , Neumonía Bacteriana/virología , Transducción de Señal/genética , Transducción de Señal/inmunología , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/virología , Staphylococcus aureus/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Colaboradores-Inductores/virología , Interleucina-22
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...