Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Nat Immunol ; 13(2): 162-9, 2012 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-22231517

RESUMEN

Memory T cells exert antigen-independent effector functions, but how these responses are regulated is unclear. We discovered an in vivo link between flagellin-induced NLRC4 inflammasome activation in splenic dendritic cells (DCs) and host protective interferon-γ (IFN-γ) secretion by noncognate memory CD8(+) T cells, which could be activated by Salmonella enterica serovar Typhimurium, Yersinia pseudotuberculosis and Pseudomonas aeruginosa. We show that CD8α(+) DCs were particularly efficient at sensing bacterial flagellin through NLRC4 inflammasomes. Although this activation released interleukin 18 (IL-18) and IL-1ß, only IL-18 was required for IFN-γ production by memory CD8(+) T cells. Conversely, only the release of IL-1ß, but not IL-18, depended on priming signals mediated by Toll-like receptors. These findings provide a comprehensive mechanistic framework for the regulation of noncognate memory T cell responses during bacterial immunity.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/inmunología , Linfocitos T CD8-positivos/inmunología , Proteínas de Unión al Calcio/inmunología , Células Dendríticas/inmunología , Memoria Inmunológica , Inflamasomas/inmunología , Interferón gamma/inmunología , Animales , Flagelina/inmunología , Interleucina-18/inmunología , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Ratones , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Salmonelosis Animal/inmunología , Salmonella typhimurium/inmunología , Transducción de Señal/inmunología , Bazo/inmunología , Receptores Toll-Like/inmunología , Infecciones por Yersinia pseudotuberculosis/inmunología
2.
J Virol ; 93(6)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30626665

RESUMEN

Airway epithelial cells and macrophages differ markedly in their responses to influenza A virus (IAV) infection. To investigate transcriptional responses underlying these differences, purified subsets of type II airway epithelial cells (ATII) and alveolar macrophages (AM) recovered from the lungs of mock- or IAV-infected mice at 9 h postinfection were subjected to RNA sequencing. This time point was chosen to allow for characterization of cell types first infected with the virus inoculum, prior to multicycle virus replication and the infiltration of inflammatory cells into the airways. In the absence of infection, AM predominantly expressed genes related to immunity, whereas ATII expressed genes consistent with their physiological roles in the lung. Following IAV infection, AM almost exclusively activated cell-intrinsic antiviral pathways that were dependent on interferon (IFN) regulatory factor 3/7 (IRF3/7) and/or type I IFN signaling. In contrast, IAV-infected ATII activated a broader range of physiological responses, including cell-intrinsic antiviral pathways, which were both independent of and dependent on IRF3/7 and/or type I IFN. These data suggest that transcriptional profiles hardwired during development are a major determinant underlying the different responses of ATII and AM to IAV infection.IMPORTANCE Airway epithelial cells (AEC) and airway macrophages (AM) represent major targets of influenza A virus (IAV) infection in the lung, yet the two cell types respond very differently to IAV infection. We have used RNA sequencing to define the host transcriptional responses in each cell type under steady-state conditions as well as following IAV infection. To do this, different cell subsets isolated from the lungs of mock- and IAV-infected mice were subjected to RNA sequencing. Under steady-state conditions, AM and AEC express distinct transcriptional activities, consistent with distinct physiological roles in the airways. Not surprisingly, these cells also exhibited major differences in transcriptional responses following IAV infection. These studies shed light on how the different transcriptional architectures of airway cells from two different lineages drive transcriptional responses to IAV infection.


Asunto(s)
Células Epiteliales/virología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Pulmón/virología , Macrófagos/virología , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/virología , Animales , Línea Celular , Enfermedades Transmisibles/metabolismo , Enfermedades Transmisibles/virología , Perros , Células Epiteliales/metabolismo , Humanos , Gripe Humana/metabolismo , Gripe Humana/virología , Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Interferones/metabolismo , Pulmón/metabolismo , Macrófagos/metabolismo , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Transducción de Señal/fisiología , Transcripción Genética/fisiología , Replicación Viral/fisiología
3.
Mol Med ; 25(1): 12, 2019 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-30943912

RESUMEN

BACKGROUND: The polymeric immunoglobulin receptor (pIgR) maintains the integrity of epithelial barriers by transporting polymeric antibodies and antigens through the epithelial mucosa into the lumen. In this study, we examined the role of pIgR in maintaining gut barrier integrity, which is important for the normal development in mice. METHODS: Cohorts of pIgR-/- mice and their wildtype controls were housed under Specific Pathogen Free (SPF) conditions and monitored for weight gain as an indicator of development over time. The general physiology of the gastrointestinal tract was analysed using immunohistochemistry in young (8-12 weeks of age) and aged mice (up to 18 months of age), and the observed immunopathology in pIgR-/- mice was further characterised using flow cytometry. Urinary metabolites were analysed using gas chromatography-mass spectrometry (GC-MS), which revealed changes in metabolites that correlated with age-related increase in gut permeability in pIgR-/- mice. RESULTS: We observed that pIgR-/- mice exhibited delayed growth, and this phenomenon is associated with low-grade gut inflammation that increased with ageing. The gross intraepithelial lymphocytic (IEL) infiltration characteristic of pIgR-/- mice was redefined as CD8α+αß+ T cells, the majority of which expressed high levels of CD103 and CD69 consistent with tissue resident memory T cells (TRM). Comparison of the urinary metabolome between pIgR-/- and wild-type mice revealed key changes in urinary biomarkers fucose, glycine and Vitamin B5, suggestive of altered mucosal permeability. A significant increase in gut permeability was confirmed by analysing the site-specific uptake of sugar probes in different parts of the intestine. CONCLUSION: Our data show that loss of the secretory antibody system in mice results in enhanced accumulation of inflammatory IELs in the gut, which likely reflects ongoing inflammation in reaction to gut microbiota or food antigens, leading to delayed growth in pIgR-/- mice. We demonstrate that this leads to the presence of a unique urinary metabolome profile, which may provide a biomarker for altered gut permeability.


Asunto(s)
Tracto Gastrointestinal/inmunología , Linfocitos Intraepiteliales/inmunología , Metaboloma , Receptores de Inmunoglobulina Polimérica/genética , Orina/química , Animales , Anticuerpos/genética , Citocinas/sangre , Femenino , Tracto Gastrointestinal/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados
4.
Blood ; 129(23): 3087-3099, 2017 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-28325862

RESUMEN

ß-Thalassemia is associated with several abnormalities of the innate immune system. Neutrophils in particular are defective, predisposing patients to life-threatening bacterial infections. The molecular and cellular mechanisms involved in impaired neutrophil function remain incompletely defined. We used the Hbbth3/+ ß-thalassemia mouse and hemoglobin E (HbE)/ß-thalassemia patients to investigate dysregulated neutrophil activity. Mature neutrophils from Hbbth3/+ mice displayed a significant reduction in chemotaxis, opsonophagocytosis, and production of reactive oxygen species, closely mimicking the defective immune functions observed in ß-thalassemia patients. In Hbbth3/+ mice, the expression of neutrophil CXCR2, CD11b, and reduced NAD phosphate oxidase components (p22phox, p67phox, and gp91phox) were significantly reduced. Morphological analysis of Hbbth3/+ neutrophils showed that a large percentage of mature phenotype neutrophils (Ly6GhiLy6Clow) appeared as band form cells, and a striking expansion of immature (Ly6GlowLy6Clow) hyposegmented neutrophils, consisting mainly of myelocytes and metamyelocytes, was noted. Intriguingly, expression of an essential mediator of neutrophil terminal differentiation, the ets transcription factor PU.1, was significantly decreased in Hbbth3/+ neutrophils. In addition, in vivo infection with Streptococcus pneumoniae failed to induce PU.1 expression or upregulate neutrophil effector functions in Hbbth3/+ mice. Similar changes to neutrophil morphology and PU.1 expression were observed in splenectomized and nonsplenectomized HbE/ß-thalassemia patients. This study provides a mechanistic insight into defective neutrophil maturation in ß-thalassemia patients, which contributes to deficiencies in neutrophil effector functions.


Asunto(s)
Neutrófilos/inmunología , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Talasemia beta/genética , Talasemia beta/inmunología , Adulto , Animales , Antígeno CD11b/metabolismo , Estudios de Casos y Controles , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Quimiotaxis de Leucocito , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Persona de Mediana Edad , Activación Neutrófila , Neutrófilos/metabolismo , Neutrófilos/patología , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/inmunología , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/inmunología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Interleucina-8B/metabolismo , Transactivadores/deficiencia , Transactivadores/inmunología , Adulto Joven , Talasemia beta/patología
5.
Clin Sci (Lond) ; 131(18): 2347-2362, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28779028

RESUMEN

Formyl peptide receptor 2/lipoxin A4 (LXA4) receptor (Fpr2/ALX) co-ordinates the transition from inflammation to resolution during acute infection by binding to distinct ligands including serum amyloid A (SAA) and Resolvin D1 (RvD1). Here, we evaluated the proresolving actions of aspirin-triggered RvD1 (AT-RvD1) in an acute coinfection pneumonia model. Coinfection with Streptococcus pneumoniae and influenza A virus (IAV) markedly increased pneumococcal lung load and neutrophilic inflammation during the resolution phase. Fpr2/ALX transcript levels were increased in the lungs of coinfected mice, and immunohistochemistry identified prominent Fpr2/ALX immunoreactivity in bronchial epithelial cells and macrophages. Levels of circulating and lung SAA were also highly increased in coinfected mice. Therapeutic treatment with exogenous AT-RvD1 during the acute phase of infection (day 4-6 post-pneumococcal inoculation) significantly reduced the pneumococcal load. AT-RvD1 also significantly reduced neutrophil elastase (NE) activity and restored total antimicrobial activity in bronchoalveolar lavage (BAL) fluid (BALF) of coinfected mice. Pneumonia severity, as measured by quantitating parenchymal inflammation or alveolitis was significantly reduced with AT-RvD1 treatment, which also reduced the number of infiltrating lung neutrophils and monocytes/macrophages as assessed by flow cytometry. The reduction in distal lung inflammation in AT-RvD1-treated mice was not associated with a significant reduction in inflammatory and chemokine mediators. In summary, we demonstrate that in the coinfection setting, SAA levels were persistently increased and exogenous AT-RvD1 facilitated more rapid clearance of pneumococci in the lungs, while concurrently reducing the severity of pneumonia by limiting excessive leukocyte chemotaxis from the infected bronchioles to distal areas of the lungs.


Asunto(s)
Aspirina/uso terapéutico , Coinfección/tratamiento farmacológico , Ácidos Docosahexaenoicos/fisiología , Infecciones por Orthomyxoviridae/complicaciones , Neumonía Neumocócica/complicaciones , Animales , Aspirina/farmacología , Carga Bacteriana/efectos de los fármacos , Modelos Animales de Enfermedad , Ácidos Docosahexaenoicos/metabolismo , Citometría de Flujo , Virus de la Influenza A , Pulmón/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Neumonía Neumocócica/tratamiento farmacológico , Receptores de Formil Péptido/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Streptococcus pneumoniae , Transcriptoma
6.
J Bacteriol ; 196(16): 3036-44, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24957617

RESUMEN

Variable-number tandem repeats (VNTRs) mutate rapidly and can be useful markers for genotyping. While multilocus VNTR analysis (MLVA) is increasingly used in the detection and investigation of food-borne outbreaks caused by Salmonella enterica serovar Typhimurium (S. Typhimurium) and other bacterial pathogens, MLVA data analysis usually relies on simple clustering approaches that may lead to incorrect interpretations. Here, we estimated the rates of copy number change at each of the five loci commonly used for S. Typhimurium MLVA, during in vitro and in vivo passage. We found that loci STTR5, STTR6, and STTR10 changed during passage but STTR3 and STTR9 did not. Relative rates of change were consistent across in vitro and in vivo growth and could be accurately estimated from diversity measures of natural variation observed during large outbreaks. Using a set of 203 isolates from a series of linked outbreaks and whole-genome sequencing of 12 representative isolates, we assessed the accuracy and utility of several alternative methods for analyzing and interpreting S. Typhimurium MLVA data. We show that eBURST analysis was accurate and informative. For construction of MLVA-based trees, a novel distance metric, based on the geometric model of VNTR evolution coupled with locus-specific weights, performed better than the commonly used simple or categorical distance metrics. The data suggest that, for the purpose of identifying potential transmission clusters for further investigation, isolates whose profiles differ at one of the rapidly changing STTR5, STTR6, and STTR10 loci should be collapsed into the same cluster.


Asunto(s)
Análisis por Conglomerados , ADN Bacteriano/genética , Repeticiones de Minisatélite , Tipificación Molecular/métodos , Tasa de Mutación , Infecciones por Salmonella/epidemiología , Salmonella typhimurium/genética , Genoma Bacteriano , Genotipo , Epidemiología Molecular/métodos , Infecciones por Salmonella/microbiología , Salmonella typhimurium/clasificación
7.
Infect Immun ; 82(1): 364-70, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24191297

RESUMEN

Otitis media (OM) (a middle ear infection) is a common childhood illness that can leave some children with permanent hearing loss. OM can arise following infection with a variety of different pathogens, including a coinfection with influenza A virus (IAV) and Streptococcus pneumoniae (the pneumococcus). We and others have demonstrated that coinfection with IAV facilitates the replication of pneumococci in the middle ear. Specifically, we used a mouse model of OM to show that IAV facilitates the outgrowth of S. pneumoniae in the middle ear by inducing middle ear inflammation. Here, we seek to understand how the host inflammatory response facilitates bacterial outgrowth in the middle ear. Using B cell-deficient infant mice, we show that antibodies play a crucial role in facilitating pneumococcal replication. We subsequently show that this is due to antibody-dependent neutrophil extracellular trap (NET) formation in the middle ear, which, instead of clearing the infection, allows the bacteria to replicate. We further demonstrate the importance of these NETs as a potential therapeutic target through the transtympanic administration of a DNase, which effectively reduces the bacterial load in the middle ear. Taken together, these data provide novel insight into how pneumococci are able to replicate in the middle ear cavity and induce disease.


Asunto(s)
Anticuerpos Antibacterianos/fisiología , Anticuerpos Antivirales/fisiología , Coinfección/microbiología , Neutrófilos/fisiología , Infecciones por Orthomyxoviridae/inmunología , Otitis Media/microbiología , Infecciones Neumocócicas/inmunología , Streptococcus pneumoniae/inmunología , Animales , Carga Bacteriana , Coinfección/virología , Modelos Animales de Enfermedad , Oído Medio/microbiología , Humanos , Virus de la Influenza A/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Orthomyxoviridae/microbiología , Otitis Media/inmunología , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/crecimiento & desarrollo
8.
Immunology ; 143(4): 520-30, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24827556

RESUMEN

Immunoglobulin A is an important mucosal antibody that can neutralize mucosal pathogens by either preventing attachment to epithelia (immune exclusion) or alternatively inhibit intra-epithelial replication following transcytosis by the polymeric immunoglobulin receptor (pIgR). Chlamydia trachomatis is a major human pathogen that initially targets the endocervical or urethral epithelium in women and men, respectively. As both tissues contain abundant secretory IgA (SIgA) we assessed the protection afforded by IgA targeting different chlamydial antigens expressed during the extra- and intra-epithelial stages of infection. We developed an in vitro model using polarizing cells expressing the murine pIgR together with antigen-specific mouse IgA, and an in vivo model using pIgR(-/-) mice. Secretory IgA targeting the extra-epithelial chlamydial antigen, the major outer membrane protein, significantly reduced infection in vitro by 24% and in vivo by 44%. Conversely, pIgR-mediated delivery of IgA targeting the intra-epithelial inclusion membrane protein A bound to the inclusion but did not reduce infection in vitro or in vivo. Similarly, intra-epithelial IgA targeting the secreted protease Chlamydia protease-like activity factor also failed to reduce infection. Together, these data suggest the importance of pIgR-mediated delivery of IgA targeting extra-epithelial, but not intra-epithelial, chlamydial antigens for protection against a genital tract infection.


Asunto(s)
Infecciones por Chlamydia/inmunología , Chlamydia/inmunología , Inmunoglobulina A Secretora/inmunología , Membrana Mucosa/inmunología , Animales , Especificidad de Anticuerpos/inmunología , Antígenos Bacterianos/inmunología , Línea Celular , Infecciones por Chlamydia/metabolismo , Chlamydia muridarum/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunoglobulina A Secretora/aislamiento & purificación , Masculino , Ratones , Ratones Noqueados , Membrana Mucosa/metabolismo , Receptores de Inmunoglobulina Polimérica/genética , Receptores de Inmunoglobulina Polimérica/metabolismo
9.
Immunogenetics ; 66(7-8): 501-6, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24906421

RESUMEN

The nonobese diabetic (NOD) mouse strain serves as a genomic standard for assessing how allelic variation for insulin-dependent diabetes (Idd) loci affects the development of autoimmune diabetes. We previously demonstrated that C57BL/6 (B6) mice harbor a more diabetogenic allele than NOD mice for the Idd14 locus when introduced onto the NOD genetic background. New congenic NOD mouse strains, harboring smaller B6-derived intervals on chromosome 13, now localize Idd14 to an ~18-Mb interval and reveal a new locus, Idd31. Notably, the B6 allele for Idd31 confers protection against diabetes, but only in the absence of the diabetogenic B6 allele for Idd14, indicating genetic epistasis between these two loci. Moreover, congenic mice that are more susceptible to diabetes are more resistant to Listeria monocytogenes infection. This result co-localizes Idd14 and Listr2, a resistance locus for listeriosis, to the same genomic interval and indicates that congenic NOD mice may also be useful for localizing resistance loci for infectious disease.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Epistasis Genética/inmunología , Listeriosis/genética , Listeriosis/inmunología , Alelos , Animales , Femenino , Predisposición Genética a la Enfermedad , Fenómenos Inmunogenéticos , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Endogámicos NOD
10.
Vaccine ; 42(7): 1714-1722, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38350767

RESUMEN

Pneumococcal Conjugate Vaccines (PCVs) have substantially reduced the burden of disease caused by Streptococcus pneumoniae (the pneumococcus). However, protection is limited to vaccine serotypes, and when administered to children who are colonized with pneumococci at the time of vaccination, immune responses to the vaccine are blunted. Here, we investigate the potential of a killed whole cell pneumococcal vaccine (WCV) to reduce existing pneumococcal carriage and mucosal disease when given therapeutically to infant mice colonized with pneumococci. We show that a single dose of WCV reduced pneumococcal carriage density in an antibody-dependent manner. Therapeutic vaccination induced robust immune responses to pneumococcal surface antigens CbpA, PspA (family 1) and PiaA. In a co-infection model of otitis media, a single dose of WCV reduced pneumococcal middle ear infection. Lastly, in a two-dose model, therapeutic administration of WCV reduced nasal shedding of pneumococci. Taken together, our data demonstrate that WCV administered in colonized mice reduced pneumococcal density in the nasopharynx and the middle ear, and decreased shedding. WCVs would be beneficial in low and middle-income settings where pneumococcal carriage in children is high.


Asunto(s)
Otitis Media , Infecciones Neumocócicas , Lactante , Niño , Humanos , Animales , Ratones , Streptococcus pneumoniae , Infecciones Neumocócicas/prevención & control , Otitis Media/prevención & control , Vacunas Neumococicas , Vacunación , Serogrupo , Vacunas Conjugadas , Nasofaringe , Portador Sano/prevención & control
11.
Infect Immun ; 81(6): 1952-63, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23509149

RESUMEN

PECAM-1/CD31 is known to regulate inflammatory responses and exhibit pro- and anti-inflammatory functions. This study was designed to determine the functional role of PECAM-1 in susceptibility to murine primary in vivo infection with Salmonella enterica serovar Typhimurium and in in vitro inflammatory responses of peritoneal macrophages. Lectin profiling showed that cellular PECAM-1 and recombinant human PECAM-1-Ig chimera contain high levels of mannose sugars and N-acetylglucosamine. Consistent with this carbohydrate pattern, both recombinant human and murine PECAM-1-Ig chimeras were shown to bind S. Typhimurium in a dose-dependent manner in vitro. Using oral and fecal-oral transmission models of S. Typhimurium SL1344 infection, PECAM-1(-/-) mice were found to be more resistant to S. Typhimurium infection than wild-type (WT) C57BL/6 mice. While fecal shedding of S. Typhimurium was comparable in wild-type and PECAM-1(-/-) mice, the PECAM-1-deficient mice had lower bacterial loads in systemic organs such as liver, spleen, and mesenteric lymph nodes than WT mice, suggesting that extraintestinal dissemination was reduced in the absence of PECAM-1. This reduced bacterial load correlated with reduced tumor necrosis factor (TNF), interleukin-6 (IL-6), and monocyte chemoattractant protein (MCP) levels in sera of PECAM-1(-/-) mice. Following in vitro stimulation of macrophages with either whole S. Typhimurium, lipopolysaccharide (LPS) (Toll-like receptor 4 [TLR4] ligand), or poly(I·C) (TLR3 ligand), production of TNF and IL-6 by PECAM-1(-/-) macrophages was reduced. Together, these results suggest that PECAM-1 may have multiple functions in resistance to infection with S. Typhimurium, including binding to host cells, extraintestinal spread to deeper tissues, and regulation of inflammatory cytokine production by infected macrophages.


Asunto(s)
Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Salmonelosis Animal/microbiología , Salmonella typhimurium/fisiología , Animales , Adhesión Bacteriana , Regulación de la Expresión Génica/inmunología , Humanos , Macrófagos Peritoneales , Ratones , Ratones Noqueados , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Proteínas Recombinantes , Salmonelosis Animal/inmunología , Salmonella typhimurium/inmunología , Organismos Libres de Patógenos Específicos , Transcriptoma
12.
Infect Immun ; 81(10): 3880-93, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23918779

RESUMEN

The natural immune response to Helicobacter pylori neither clears infection nor prevents reinfection. However, the ability of secretory antibodies to influence the course of H. pylori infection has not been determined. We compared the natural progression of H. pylori infection in wild-type C57BL/6 mice with that in mice lacking the polymeric immunoglobulin receptor (pIgR) that is essential for the secretion of polymeric antibody across mucosal surfaces. H. pylori SS1-infected wild-type and pIgR knockout (KO) mice were sampled longitudinally for gastrointestinal bacterial load, antibody response, and histological changes. The gastric bacterial loads of wild-type and pIgR KO mice remained constant and comparable at up to 3 months postinfection (mpi) despite SS1-reactive secretory IgA in the intestinal contents of wild-type mice at that time. Conversely, abundant duodenal colonization of pIgR KO animals contrasted with the near-total eradication of H. pylori from the intestine of wild-type animals by 3 mpi. H. pylori was cultured only from the duodenum of those animals in which colonization in the distal gastric antrum was of sufficient density for immunohistological detection. By 6 mpi, the gastric load of H. pylori in wild-type mice was significantly lower than in pIgR KO animals. While there was no corresponding difference between the two mouse strains in gastric pathology results at 6 mpi, reductions in gastric bacterial load correlated with increased gastric inflammation together with an intestinal secretory antibody response in wild-type mice. Together, these results suggest that naturally produced secretory antibodies can modulate the progress of H. pylori infection, particularly in the duodenum.


Asunto(s)
Anticuerpos Antibacterianos/metabolismo , Infecciones por Helicobacter/inmunología , Helicobacter pylori , Inmunidad Mucosa/fisiología , Mucosa Intestinal/metabolismo , Animales , Western Blotting , Regulación de la Expresión Génica/inmunología , Inmunoglobulina A/sangre , Inmunoglobulina A/metabolismo , Inmunoglobulina G/sangre , Inmunoglobulina G/metabolismo , Mucosa Intestinal/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Inmunoglobulina Polimérica/genética , Receptores de Inmunoglobulina Polimérica/metabolismo
13.
Infect Immun ; 81(3): 645-52, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23319557

RESUMEN

Influenza A virus (IAV) predisposes individuals to secondary infections with the bacterium Streptococcus pneumoniae (the pneumococcus). Infections may manifest as pneumonia, sepsis, meningitis, or otitis media (OM). It remains controversial as to whether secondary pneumococcal disease is due to the induction of an aberrant immune response or IAV-induced immunosuppression. Moreover, as the majority of studies have been performed in the context of pneumococcal pneumonia, it remains unclear how far these findings can be extrapolated to other pneumococcal disease phenotypes such as OM. Here, we used an infant mouse model, human middle ear epithelial cells, and a series of reverse-engineered influenza viruses to investigate how IAV promotes bacterial OM. Our data suggest that the influenza virus HA facilitates disease by inducing a proinflammatory response in the middle ear cavity in a replication-dependent manner. Importantly, our findings suggest that it is the inflammatory response to IAV infection that mediates pneumococcal replication. This study thus provides the first evidence that inflammation drives pneumococcal replication in the middle ear cavity, which may have important implications for the treatment of pneumococcal OM.


Asunto(s)
Inflamación/patología , Infecciones por Orthomyxoviridae/complicaciones , Otitis Media/patología , Infecciones Neumocócicas/patología , Animales , Virus de la Influenza A/clasificación , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/microbiología , Infecciones por Orthomyxoviridae/virología , Otitis Media/inmunología , Otitis Media/microbiología , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/microbiología , Carga Viral
14.
PLoS Pathog ; 7(8): e1002204, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21901098

RESUMEN

Klebsiella pneumoniae causes significant morbidity and mortality worldwide, particularly amongst hospitalized individuals. The principle mechanism for pathogenesis in hospital environments involves the formation of biofilms, primarily on implanted medical devices. In this study, we constructed a transposon mutant library in a clinical isolate, K. pneumoniae AJ218, to identify the genes and pathways implicated in biofilm formation. Three mutants severely defective in biofilm formation contained insertions within the mrkABCDF genes encoding the main structural subunit and assembly machinery for type 3 fimbriae. Two other mutants carried insertions within the yfiN and mrkJ genes, which encode GGDEF domain- and EAL domain-containing c-di-GMP turnover enzymes, respectively. The remaining two isolates contained insertions that inactivated the mrkH and mrkI genes, which encode for novel proteins with a c-di-GMP-binding PilZ domain and a LuxR-type transcriptional regulator, respectively. Biochemical and functional assays indicated that the effects of these factors on biofilm formation accompany concomitant changes in type 3 fimbriae expression. We mapped the transcriptional start site of mrkA, demonstrated that MrkH directly activates transcription of the mrkA promoter and showed that MrkH binds strongly to the mrkA regulatory region only in the presence of c-di-GMP. Furthermore, a point mutation in the putative c-di-GMP-binding domain of MrkH completely abolished its function as a transcriptional activator. In vivo analysis of the yfiN and mrkJ genes strongly indicated their c-di-GMP-specific function as diguanylate cyclase and phosphodiesterase, respectively. In addition, in vitro assays showed that purified MrkJ protein has strong c-di-GMP phosphodiesterase activity. These results demonstrate for the first time that c-di-GMP can function as an effector to stimulate the activity of a transcriptional activator, and explain how type 3 fimbriae expression is coordinated with other gene expression programs in K. pneumoniae to promote biofilm formation to implanted medical devices.


Asunto(s)
Biopelículas , GMP Cíclico/análogos & derivados , Fimbrias Bacterianas/metabolismo , Klebsiella pneumoniae/genética , Activación Transcripcional , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , GMP Cíclico/genética , ADN Bacteriano/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Fimbrias Bacterianas/genética , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica , Klebsiella pneumoniae/crecimiento & desarrollo , Klebsiella pneumoniae/metabolismo , Datos de Secuencia Molecular , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Liasas de Fósforo-Oxígeno/genética , Liasas de Fósforo-Oxígeno/metabolismo , Plásmidos , Unión Proteica
15.
Virol J ; 10: 128, 2013 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-23617940

RESUMEN

BACKGROUND: Otitis media (OM) affects ≥80% of children before the age of three. OM can arise following co-infection with influenza A virus (IAV) and the bacterium Streptococcus pneumoniae. We have previously shown that H3 IAV strains (such as Udorn/72) induced a higher rate of bacterial OM than H1 strains (such as PR8/34). This was associated with more efficient replication of H3 strains in the middle ear. FINDINGS: Here, we assess if the increased replication of IAV strains such as Udorn/72 in the middle ear is dependent upon the binding of the viral HA to α2,6-linked sialic acid. Using murine and in vitro models, the present study shows that recognition of α2,6-linked sialic acid was not required to facilitate bacterial OM. CONCLUSIONS: Taken together, these data suggest that other features of the HA mediate bacterial OM.


Asunto(s)
Virus de la Influenza A/fisiología , Ácido N-Acetilneuramínico/metabolismo , Infecciones por Orthomyxoviridae/complicaciones , Otitis Media/patología , Infecciones Neumocócicas/patología , Tropismo Viral , Acoplamiento Viral , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/virología , Otitis Media/microbiología , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/patogenicidad , Replicación Viral
16.
J Exp Med ; 203(1): 21-6, 2006 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-16390940

RESUMEN

The production of IgA is induced in an antigen-unspecific manner by commensal flora. These secretory antibodies (SAbs) may bind multiple antigens and are thought to eliminate commensal bacteria and self-antigens to avoid systemic recognition. In this study, we addressed the role of "innate" SAbs, i.e., those that are continuously produced in normal individuals, in protection against infection of the gastrointestinal tract. We used polymeric immunoglobulin receptor (pIgR-/-) knock-out mice, which are unable to bind and actively transport dimeric IgA and pentameric IgM to the mucosae, and examined the role of innate SAbs in protection against the invasive pathogen Salmonella typhimurium. In vitro experiments suggested that innate IgA in pIgR-/- serum bound S. typhimurium in a cross-reactive manner which inhibited epithelial cell invasion. Using a "natural" infection model, we demonstrated that pIgR-/- mice are profoundly sensitive to infection with S. typhimurium via the fecal-oral route and, moreover, shed more bacteria that readily infected other animals. These results imply an important evolutionary role for innate SAbs in protecting both the individual and the herd against infections, and suggest that the major role of SAbs may be to prevent the spread of microbial pathogens throughout the population, rather than protection of local mucosal surfaces.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Receptores de Inmunoglobulina Polimérica/deficiencia , Salmonelosis Animal/inmunología , Salmonella typhimurium/patogenicidad , Animales , Línea Celular , Recuento de Colonia Microbiana , Perros , Heces/microbiología , Inmunidad Innata , Inmunoglobulina A/sangre , Intestino Delgado/inmunología , Intestino Delgado/microbiología , Dosificación Letal Mediana , Ratones , Ratones Endogámicos , Ratones Noqueados , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/microbiología , Receptores de Inmunoglobulina Polimérica/sangre , Receptores de Inmunoglobulina Polimérica/genética , Salmonelosis Animal/mortalidad , Salmonelosis Animal/transmisión
17.
J Immunol ; 184(5): 2237-42, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20164434

RESUMEN

Given the central role of intestinal dendritic cells (DCs) in the regulation of gut immune responses, it is not surprising that several bacterial pathogens have evolved strategies to prevent or bypass recognition by DCs. In this article, we will review recent findings on the interaction between intestinal DCs and prototypical bacterial pathogens, such as Salmonella, Yersinia, or Helicobacter. We will discuss the different approaches with which these pathogens seek to evade DC recognition and subsequent T cell activation. These diverse strategies span to include mounting irrelevant immune responses, inhibition of Ag presentation by DCs, and stretch as far as to manipulate the Th1/Th2 balance of CD4(+) T cells in the bacteria's favor.


Asunto(s)
Células Dendríticas/inmunología , Helicobacter pylori/inmunología , Intestinos/inmunología , Salmonella typhimurium/inmunología , Yersinia enterocolitica/inmunología , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Helicobacter pylori/fisiología , Interacciones Huésped-Patógeno/inmunología , Humanos , Intestinos/microbiología , Modelos Inmunológicos , Salmonella typhimurium/fisiología , Yersinia enterocolitica/fisiología
18.
J Infect Dis ; 204(12): 1857-65, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21930608

RESUMEN

Otitis media (OM) is one of the most common childhood diseases. OM can arise when a viral infection enables bacteria to disseminate from the nasopharynx to the middle ear. Here, we provide the first infant murine model for disease. Mice coinfected with Streptococcus pneumoniae and influenza virus had high bacterial load in the middle ear, middle ear inflammation, and hearing loss. In contrast, mice colonized with S. pneumoniae alone had significantly less bacteria in the ear, minimal hearing loss, and no inflammation. Of interest, infection with influenza virus alone also caused some middle ear inflammation and hearing loss. Overall, this study provides a clinically relevant and easily accessible animal model to study the pathogenesis and prevention of OM. Moreover, we provide, to our knowledge, the first evidence that influenza virus alone causes middle ear inflammation in infant mice. This inflammation may then play an important role in the development of bacterial OM.


Asunto(s)
Potenciales Evocados Auditivos del Tronco Encefálico , Virus de la Influenza A , Infecciones por Orthomyxoviridae/complicaciones , Otitis Media/microbiología , Streptococcus pneumoniae , Animales , Carga Bacteriana , Coinfección , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Mediciones Luminiscentes , Ratones , Ratones Endogámicos C57BL , Otitis Media/patología , Otitis Media/fisiopatología , Otitis Media/virología
19.
mSphere ; 7(1): e0098421, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35044807

RESUMEN

Streptococcus pneumoniae (the pneumococcus) is a leading cause of pneumonia in children under 5 years of age. Coinfection by pneumococci and respiratory viruses enhances disease severity. Little is known about pneumococcal coinfections with respiratory syncytial virus (RSV). Here, we developed a novel infant mouse model of coinfection using pneumonia virus of mice (PVM), a murine analogue of RSV, to examine the dynamics of coinfection in the upper respiratory tract, an anatomical niche that is essential for host-to-host transmission and progression to disease. Coinfection increased damage to the nasal tissue and increased production of the chemokine CCL3. Nasopharyngeal pneumococcal density and shedding in nasal secretions were increased by coinfection. In contrast, coinfection reduced PVM loads in the nasopharynx, an effect that was independent of pneumococcal strain and the order of infection. We showed that this "antagonistic" effect was absent using either ethanol-killed pneumococci or a pneumococcal mutant deficient in capsule production and incapable of nasopharyngeal carriage. Colonization with a pneumococcal strain naturally unable to produce capsule also reduced viral loads. The pneumococcus-mediated reduction in PVM loads was caused by accelerated viral clearance from the nasopharynx. Although these synergistic and antagonistic effects occurred with both wild-type pneumococcal strains used in this study, the magnitude of the effects was strain dependent. Lastly, we showed that pneumococci can also antagonize influenza virus. Taken together, our study has uncovered multiple novel facets of bacterial-viral coinfection. Our findings have important public health implications, including for bacterial and viral vaccination strategies in young children. IMPORTANCE Respiratory bacterial-viral coinfections (such as pneumococci and influenza virus) are often synergistic, resulting in enhanced disease severity. Although colonization of the nasopharynx is the precursor to disease and transmission, little is known about bacterial-viral interactions that occur within this niche. In this study, we developed a novel mouse model to examine pneumococcal-viral interactions in the nasopharynx with pneumonia virus of mice (PVM) and influenza. We found that PVM infection benefits pneumococci by increasing their numbers in the nasopharynx and shedding of these bacteria in respiratory secretions. In contrast, we discovered that pneumococci decrease PVM numbers by accelerating viral clearance. We also report a similar effect of pneumococci on influenza. By showing that coinfections lead to both synergistic and antagonistic outcomes, our findings challenge the existing dogma in the field. Our work has important applications and implications for bacterial and viral vaccines that target these microbes.


Asunto(s)
Antibiosis , Coinfección/microbiología , Coinfección/virología , Infecciones Neumocócicas/virología , Infecciones por Pneumovirus/virología , Sistema Respiratorio/virología , Factores de Edad , Animales , Coinfección/inmunología , Citocinas/análisis , Citocinas/inmunología , Modelos Animales de Enfermedad , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , Ratones , Ratones Endogámicos C57BL , Virus de la Neumonía Murina/genética , Virus de la Neumonía Murina/inmunología , Nasofaringe/virología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Infecciones por Pneumovirus/inmunología , Sistema Respiratorio/inmunología , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/inmunología , Carga Viral
20.
Eur J Immunol ; 40(10): 2778-90, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21038469

RESUMEN

Helicobacter pylori is recognised as the chief cause of chronic gastritis, ulcers and gastric cancer in humans. With increased incidence of treatment failure and antibiotic resistance, development of prophylactic or therapeutic vaccination is a desirable alternative. Although the results of vaccination studies in animal models have been promising, studies in human volunteers have revealed problems such as 'post-immunisation gastritis' and comparatively poor responses to vaccine antigens. The focus of this study was to compare the gastric and systemic cellular immune responses induced by recombinant attenuated Salmonella Typhimurium-based vaccination in the C57BL/6 model of H. pylori infection. Analysis of lymphocyte populations in the gastric mucosa, blood, spleen, paragastric LN and MLN revealed that the effects of vaccination were largely confined to the parenchymal stomach rather than lymphoid organs. Vaccine-induced protection was correlated with an augmented local recall response in the gastric mucosa, with increased proportions of CD4(+) T cells, neutrophils and reduced proportions of CD4(+) Treg. CD4(+) T cells isolated from the stomachs of vaccinated mice proliferated ex vivo in response to H. pylori antigen, and secreted Th1 cytokines, particularly IFN-γ. This detailed analysis of local gastric immune responses provides insight into the mechanism of vaccine-induced protection.


Asunto(s)
Vacunas Bacterianas/inmunología , Gastritis/microbiología , Gastritis/prevención & control , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/prevención & control , Helicobacter pylori/inmunología , Vacunación/métodos , Animales , Vacunas Bacterianas/administración & dosificación , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Femenino , Citometría de Flujo , Mucosa Gástrica/inmunología , Mucosa Gástrica/microbiología , Gastritis/inmunología , Infecciones por Helicobacter/microbiología , Histocitoquímica , Memoria Inmunológica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Estadísticas no Paramétricas , Células TH1/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda