Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros

Bases de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Infect Immun ; 81(1): 381-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23147039

RESUMEN

The respiratory epithelium is a physical and functional barrier actively involved in the clearance of environmental agents. The alveolar compartment is lined with membranous pneumocytes, known as type I alveolar epithelial cells (AEC I), and granular pneumocytes, type II alveolar epithelial cells (AEC II). AEC II are responsible for epithelial reparation upon injury and ion transport and are very active immunologically, contributing to lung defense by secreting antimicrobial factors. AEC II also secrete a broad variety of factors, such as cytokines and chemokines, involved in activation and differentiation of immune cells and are able to present antigen to specific T cells. Another cell type important in lung defense is the pulmonary macrophage (PuM). Considering the architecture of the alveoli, a good communication between the external and the internal compartments is crucial to mount effective responses. Our hypothesis is that being in the interface, AEC may play an important role in transmitting signals from the external to the internal compartment and in modulating the activity of PuM. For this, we collected supernatants from AEC unstimulated or stimulated in vitro with lipopolysaccharide (LPS). These AEC-conditioned media were used in various setups to test for the effects on a number of macrophage functions: (i) migration, (ii) phagocytosis and intracellular control of bacterial growth, and (iii) phenotypic changes and morphology. Finally, we tested the direct effect of AEC-conditioned media on bacterial growth. We found that AEC-secreted factors had a dual effect, on one hand controlling bacterial growth and on the other hand increasing macrophage activity.


Asunto(s)
Infecciones Bacterianas/metabolismo , Células Epiteliales/metabolismo , Macrófagos Alveolares/metabolismo , Alveolos Pulmonares/metabolismo , Mucosa Respiratoria/metabolismo , Sistema Respiratorio/metabolismo , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Comunicación Celular/inmunología , Movimiento Celular/inmunología , Células Cultivadas , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Femenino , Lipopolisacáridos/inmunología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/microbiología , Ratones , Ratones Endogámicos C57BL , Fagocitosis/inmunología , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/microbiología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/microbiología , Sistema Respiratorio/inmunología , Cicatrización de Heridas/inmunología
2.
Infect Immun ; 79(11): 4649-56, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21844233

RESUMEN

In the present study, we addressed the question of whether Toll-like receptor 2 (TLR2)-mediated innate immunity can contribute to the development of acquired immune responses. We immunized TLR2(-/-) and wild-type (WT) mice three times subcutaneously with the mycobacterial antigen (Ag19kDa) (a TLR2 ligand) or Ag85A (not a TLR2 ligand). One week after the last immunization, sera and spleens were collected. To evaluate cellular responses, we measured gamma interferon (IFN-γ) after in vitro restimulation of spleen cells with antigen alone or antigen-pulsed bone marrow-derived macrophages (BMM(Ag)) or pulmonary macrophages (PuM(Ag)). Antibody responses were comparable in the two mouse strains, but we observed differences in the cellular responses. Recall responses to Ag85A were similar in the two strains, but responses to Ag19kDa given alone or presented by BMM or PuM were lower in TLR2(-/-) than in WT mice. The largest differences in cellular responses were observed when Ag19kDa was presented by PuM. To understand this, we analyzed phenotypic and functional differences between BMM and PuM upon stimulation with various ligands. Generally, PuM had a lower response to the TLR2 ligand Pam(3)Cys-Ser-(Lys)(4) trihydrochloride and to anti-CD40 than BMM, as measured by cytokine secretion and upregulation of costimulatory molecules. This might provide a partial explanation for the lower capacity of PuM when pulsed with Ag19kDa, also a TLR2 ligand. Altogether, our results revealed weaknesses in the T cell and antigen-presenting cell (APC) compartments of the Ag19kDa-immunized TLR2(-/-) mice but indicated that specific immune responses could be generated in the absence of TLR2 regardless of the characteristics of the antigen used.


Asunto(s)
Antígenos Bacterianos/inmunología , Mycobacterium/inmunología , Receptor Toll-Like 2/metabolismo , Animales , Interferón gamma/genética , Interferón gamma/metabolismo , Macrófagos/metabolismo , Macrófagos Alveolares/metabolismo , Ratones , Ratones Noqueados , Mycobacterium/metabolismo , Bazo/citología , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética
3.
Pathog Dis ; 76(8)2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30476069

RESUMEN

Bacterial infection of the kidney leads to a rapid cascade of host protective responses, many of which are still poorly understood. We have previously shown that following kidney infection with uropathogenic Escherichia coli (UPEC), vascular coagulation is quickly initiated in local perivascular capillaries that protects the host from progressing from a local infection to systemic sepsis. The signaling mechanisms behind this response have not however been described. In this study, we use a number of in vitro and in vivo techniques, including intravital microscopy, to identify two previously unrecognized components influencing this protective coagulation response. The acylation state of the Lipid A of UPEC lipopolysaccharide (LPS) is shown to alter the kinetics of local coagulation onset in vivo. We also identify epithelial CD147 as a potential host factor influencing infection-mediated coagulation. CD147 is expressed by renal proximal epithelial cells infected with UPEC, contingent to bacterial expression of the α-hemolysin toxin. The epithelial CD147 subsequently can activate tissue factor on endothelial cells, a primary step in the coagulation cascade. This study emphasizes the rapid, multifaceted response of the kidney tissue to bacterial infection and the interplay between host and pathogen during the early hours of renal infection.


Asunto(s)
Infecciones Bacterianas/sangre , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/metabolismo , Basigina/metabolismo , Coagulación Sanguínea , Lípido A/inmunología , Nefritis/etiología , Nefritis/metabolismo , Animales , Biomarcadores , Línea Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Proteoma , Proteómica/métodos , Ratas , Transducción de Señal
4.
PLoS One ; 9(8): e103411, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25089618

RESUMEN

Tissue-resident macrophages are heterogeneous with tissue-specific and niche-specific functions. Thus, simplified models of macrophage activation do not explain the extent of heterogeneity seen in vivo. We focus here on the respiratory tract and ask whether factors secreted by alveolar epithelial cells (AEC) can influence the functionality of resident pulmonary macrophages (PuM). We have previously reported that factors secreted by AEC increase control of intracellular growth of BCG in macrophages. In the current study, we also aimed to investigate possible mechanisms by which AEC-derived factors increase intracellular control of BCG in both primary murine interstitial macrophages, and bone marrow-derived macrophages and characterize further the effect of these factors on macrophage differentiation. We show that; a) in contrast to other macrophage types, IFN-γ did not increase intracellular growth control of Mycobacterium bovis, Bacillus Calmette-Guérin (BCG) by interstitial pulmonary macrophages although the same macrophages could be activated by factors secreted by AEC; b) the lack of response of pulmonary macrophages to IFN-γ was apparently regulated by suppressor of cytokine signaling (SOCS)1; c) AEC-derived factors did not induce pro-inflammatory pathways induced by IFN-γ e.g. expression of inducible nitric oxide synthase (iNOS), secretion of nitric oxide (NO), or IL-12, d) in contrast to IFN-γ, intracellular bacterial destruction induced by AEC-derived factors was not dependent on iNOS transcription and NO production. Collectively, our data show that PuM were restricted in inflammatory responses mediated by IFN-γ through SOCS1 and that factors secreted by AEC- enhanced the microbicidal capacities of macrophages by iNOS independent mechanisms.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Macrófagos/metabolismo , Mycobacterium/fisiología , Óxido Nítrico/metabolismo , Fagocitosis , Células Epiteliales Alveolares/efectos de los fármacos , Animales , Células de la Médula Ósea/citología , Cloroquina/farmacología , Femenino , Interferón gamma/farmacología , Interleucina-1beta/genética , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/microbiología , Macrófagos/efectos de los fármacos , Ratones Endogámicos C57BL , Mycobacterium/efectos de los fármacos , Mycobacterium/crecimiento & desarrollo , Mycobacterium bovis/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fagocitosis/efectos de los fármacos , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Estallido Respiratorio/efectos de los fármacos , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Transcripción Genética/efectos de los fármacos
5.
PLoS One ; 8(7): e69287, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23861965

RESUMEN

Non-hematopoietic cells, including lung epithelial cells, influence host immune responses. By co-culturing primary alveolar epithelial cells and monocytes from naïve donor mice, we show that alveolar epithelial cells support monocyte survival and differentiation in vitro, suggesting a role for non-hematopoietic cells in monocyte differentiation during the steady state in vivo. CD103(+) dendritic cells (αE-DC) are present at mucosal surfaces. Using a murine primary monocyte adoptive transfer model, we demonstrate that αE-DC in the lungs and pulmonary lymph nodes are monocyte-derived during pulmonary tuberculosis. The tissue localization may influence the functional potential of αE-DC that accumulate in Mycobacterium tuberculosis-infected lungs. Here, we confirm the localization of αE-DC in uninfected mice beneath the bronchial epithelial cell layer and near the vascular wall, and show that αE-DC have a similar distribution in the lungs during pulmonary tuberculosis and are detected in the bronchoalveolar lavage fluid from infected mice. Lung DC can be targeted by M. tuberculosis in vivo and play a role in bacterial dissemination to the draining lymph node. In contrast to other DC subsets, only a fraction of lung αE-DC are infected with the bacterium. We also show that virulent M. tuberculosis does not significantly alter cell surface expression levels of MHC class II on infected cells in vivo and that αE-DC contain the highest frequency of IL-12p40(+) cells among the myeloid cell subsets in infected lungs. Our results support a model in which inflammatory monocytes are recruited into the M. tuberculosis-infected lung tissue and, depending on which non-hematopoietic cells they interact with, differentiate along different paths to give rise to multiple monocyte-derived cells, including DC with a distinctive αE-DC phenotype.


Asunto(s)
Antígenos CD/metabolismo , Células Dendríticas/patología , Cadenas alfa de Integrinas/metabolismo , Subunidad p40 de la Interleucina-12/biosíntesis , Pulmón/patología , Monocitos/patología , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/patología , Células Epiteliales Alveolares/microbiología , Células Epiteliales Alveolares/patología , Animales , Bronquios/microbiología , Bronquios/patología , Recuento de Células , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Células Dendríticas/inmunología , Regulación hacia Abajo , Femenino , Antígenos de Histocompatibilidad Clase II/metabolismo , Inflamación/patología , Pulmón/inmunología , Pulmón/microbiología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/fisiología , Tuberculosis Pulmonar/microbiología
6.
PLoS One ; 7(2): e32125, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22393384

RESUMEN

Macrophages and dendritic cells have been recognized as key players in the defense against mycobacterial infection. However, more recently, other cells in the lungs such as alveolar epithelial cells (AEC) have been found to play important roles in the defense and pathogenesis of infection. In the present study we first compared AEC with pulmonary macrophages (PuM) isolated from mice in their ability to internalize and control Bacillus Calmette-Guérin (BCG) growth and their capacity as APCs. AEC were able to internalize and control bacterial growth as well as present antigen to primed T cells. Secondly, we compared both cell types in their capacity to secrete cytokines and chemokines upon stimulation with various molecules including mycobacterial products. Activated PuM and AEC displayed different patterns of secretion. Finally, we analyzed the profile of response of AEC to diverse stimuli. AEC responded to both microbial and internal stimuli exemplified by TLR ligands and IFNs, respectively. The response included synthesis by AEC of several factors, known to have various effects in other cells. Interestingly, TNF could stimulate the production of CCL2/MCP-1. Since MCP-1 plays a role in the recruitment of monocytes and macrophages to sites of infection and macrophages are the main producers of TNF, we speculate that both cell types can stimulate each other. Also, another cell-cell interaction was suggested when IFNs (produced mainly by lymphocytes) were able to induce expression of chemokines (IP-10 and RANTES) by AEC involved in the recruitment of circulating lymphocytes to areas of injury, inflammation, or viral infection. In the current paper we confirm previous data on the capacity of AEC regarding internalization of mycobacteria and their role as APC, and extend the knowledge of AEC as a multifunctional cell type by assessing the secretion of a broad array of factors in response to several different types of stimuli.


Asunto(s)
Inmunidad Adaptativa , Células Epiteliales/citología , Inmunidad Innata , Alveolos Pulmonares/citología , Animales , Presentación de Antígeno , Quimiocina CCL2/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Femenino , Citometría de Flujo/métodos , Sistema Inmunológico , Pulmón/inmunología , Macrófagos Alveolares/citología , Macrófagos Alveolares/inmunología , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Mycobacterium/metabolismo , Mycobacterium bovis/metabolismo
7.
Vaccine ; 27(52): 7418-27, 2009 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-19735756

RESUMEN

In this study, we have compared the immunological responses associated with early pulmonary mycobacterial infection in two mouse strains, BALB/c and C57BL/6 known to exhibit distinct differences in susceptibility to infection with several pathogens. We infected mice via the intranasal route. We have demonstrated that BALB/c was less able to control mycobacterial growth in the lungs during the early phase of pulmonary infection. Our results showed that during the early phase (day 3 to week 1), BALB/c mice exhibited a delay in the production of TNF and IFN-gamma in the lungs compared to C57BL/6 mice. Levels of IL-12 and soluble TNF receptors (sTNFR) were comparable between the mouse strains. The cellular subset distribution in these mice before and after infection showed a higher increase in CD11b+ cells in the lungs of C57BL/6, compared to BALB/c as early as day 3 postinfection. At early time points, higher levels of monocyte chemoattractant protein (MCP)-1 and macrophage inflammatory protein 1 (MIP)-alpha were detected in C57BL/6 than BALB/c mice. In vitro, BCG-infected bone marrow derived macrophages (BMM) from both mouse strains displayed similar capacities to either phagocytose bacteria or produce soluble mediators such as TNF, IL-12 and nitric oxide (NO). Although IFN-gamma stimulation of infected BMM in both mouse strains resulted in the induction of antimycobacterial activity, BALB/c mice had a reduced capacity to kill ingested bacteria. The above observations indicate that the chain of early, possibly innate immunological events occurring during pulmonary mycobacterial infection may directly impact on increased susceptibility or resistance to infection.


Asunto(s)
Inmunidad Innata , Macrófagos/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Quimiocina CCL2/inmunología , Quimiocina CCL3/inmunología , Femenino , Interferón gamma , Interleucina-12/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Óxido Nítrico/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA