Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Nat Immunol ; 13(9): 843-50, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22863752

RESUMEN

A large gap in our understanding of infant immunity is why natural killer (NK) cell responses are deficient, which makes infants more prone to viral infection. Here we demonstrate that transforming growth factor-ß (TGF-ß) was responsible for NK cell immaturity during infancy. We found more fully mature NK cells in CD11c(dnR) mice, whose NK cells lack TGF-ß receptor (TGF-ßR) signaling. Ontogenic maturation of NK cells progressed faster in the absence of TGF-ß signaling, which results in the formation of a mature NK cell pool early in life. As a consequence, infant CD11c(dnR) mice efficiently controlled viral infections. These data thus demonstrate an unprecedented role for TGF-ß in ontogeny that can explain why NK cell responses are deficient early in life.


Asunto(s)
Sistema Inmunológico/citología , Sistema Inmunológico/crecimiento & desarrollo , Células Asesinas Naturales/citología , Factor de Crecimiento Transformador beta/inmunología , Animales , Animales Recién Nacidos , Diferenciación Celular/inmunología , Citometría de Flujo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Madre , Factor de Crecimiento Transformador beta/metabolismo
2.
Am J Physiol Lung Cell Mol Physiol ; 324(4): L536-L549, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36852927

RESUMEN

Interstitial macrophages (IMs) reside in the lung tissue surrounding key structures including airways, vessels, and alveoli. Recent work has described IM heterogeneity during homeostasis, however, there are limited data on IMs during inflammation. We sought to characterize IM origin, subsets, and transcriptomic profiles during homeostasis and lipopolysaccharide (LPS) induced acute lung inflammation. During homeostasis, we used three complementary methods, spectral flow cytometry, single-cell RNA-sequencing, and gene regulatory network enrichment, to demonstrate that IMs can be divided into two core subsets distinguished by surface and transcriptional expression of folate receptor ß (Folr2/FRß). These subsets inhabited distinct niches within the lung interstitium. Within FRß+ IMs we identified a subpopulation marked by coexpression of LYVE1. During acute LPS-induced inflammation, lung IM numbers expand. Lineage tracing revealed IM expansion was due to recruitment of monocyte-derived IMs. At the peak of inflammation, recruited IMs were comprised two unique subsets defined by expression of genes associated with interferon signaling and glycolytic pathways. As recruited IMs matured, they adopted the overall transcriptional state of FRß- resident IMs but retained expression in several origin-specific genes, such as IL-1ß. FRß+ IMs were of near-pure resident origin. Taken together our data show that during LPS-induced inflammation, there are distinct populations of IMs that likely have unique functions. FRΒ+ IMs comprise a stable, resident population, whereas FRß- ΙΜs represent a mixed population of resident and recruited IMs.


Asunto(s)
Receptor 2 de Folato , Neumonía , Humanos , Monocitos/metabolismo , Lipopolisacáridos/farmacología , Lipopolisacáridos/metabolismo , Macrófagos/metabolismo , Neumonía/inducido químicamente , Neumonía/genética , Neumonía/metabolismo , Inflamación/genética , Inflamación/metabolismo , Análisis de Secuencia de ARN/métodos , Receptor 2 de Folato/metabolismo
3.
Am J Physiol Lung Cell Mol Physiol ; 323(4): L391-L399, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35943156

RESUMEN

The pathogenesis of chronic obstructive pulmonary disease (COPD), a prevalent disease primarily caused by cigarette smoke exposure, is incompletely elucidated. Studies in humans and mice have suggested that hypoxia-inducible factor-1α (HIF-1α) may play a role. Reduced lung levels of HIF-1α are associated with decreased vascular density, whereas increased leukocyte HIF-1α may be responsible for increased inflammation. To elucidate the specific role of leukocyte HIF-1α in COPD, we exposed transgenic mice with conditional deletion or overexpression of HIF-1α in leukocytes to cigarette smoke for 7 mo. Outcomes included pulmonary physiology, aerated lung volumes via microcomputed tomography, lung morphometry and histology, and cardiopulmonary hemodynamics. On aggregate, cigarette smoke increased the aerated lung volume, quasi-static lung compliance, inspiratory capacity of all strains while reducing the total alveolar septal volume. Independent of smoke exposure, mice with leukocyte-specific HIF-1α overexpression had increased quasi-static compliance, inspiratory capacity, and alveolar septal volume compared with mice with leukocyte-specific HIF-1α deletion. However, the overall development of cigarette smoke-induced lung disease did not vary relative to control mice for either of the conditional strains. This suggests that the development of murine cigarette smoke-induced airspace disease occurs independently of leukocyte HIF-1α signaling.


Asunto(s)
Fumar Cigarrillos , Enfermedad Pulmonar Obstructiva Crónica , Enfisema Pulmonar , Animales , Modelos Animales de Enfermedad , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Leucocitos , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfisema Pulmonar/patología , Nicotiana/efectos adversos , Microtomografía por Rayos X
4.
Am J Respir Crit Care Med ; 203(8): 946-956, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33079572

RESUMEN

Rationale: Macrophages are the most abundant immune cell in the alveoli and small airways and are traditionally viewed as a homogeneous population during health. Whether distinct subsets of airspace macrophages are present in healthy humans is unknown. Single-cell RNA sequencing allows for examination of transcriptional heterogeneity between cells and between individuals. Understanding the conserved repertoire of airspace macrophages during health is essential to understanding cellular programing during disease.Objectives: We sought to determine the transcriptional heterogeneity of human cells obtained from BAL of healthy adults.Methods: Ten subjects underwent bronchoscopy with BAL. Cells from lavage were subjected to single-cell RNA sequencing. Unique cell populations and putative functions were identified. Transcriptional profiles were compared across individuals.Measurements and Main Results: We identify two novel subgroups of resident airspace macrophages-defined by proinflammatory and metallothionein gene expression profiles. We define subsets of monocyte-like cells and compare them with peripheral blood mononuclear cells. Finally, we compare global macrophage and monocyte programing between males and females.Conclusions: Healthy human airspaces contain multiple populations of myeloid cells that are highly conserved between individuals and between sexes. Resident macrophages make up the largest population and include novel subsets defined by inflammatory and metal-binding profiles. Monocyte-like cells within the airspaces are transcriptionally aligned with circulating blood cells and include a rare population defined by expression of cell-matrix interaction genes. This study is the first to delineate the conserved heterogeneity of airspace immune cells during health and identifies two previously unrecognized macrophage subsets.


Asunto(s)
Líquido del Lavado Bronquioalveolar/inmunología , Perfilación de la Expresión Génica , Leucocitos Mononucleares/inmunología , Macrófagos Alveolares/inmunología , Monocitos/inmunología , Alveolos Pulmonares/inmunología , Análisis de Secuencia de ARN , Adulto , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Factores Sexuales
5.
Am J Respir Cell Mol Biol ; 64(5): 629-640, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33662226

RESUMEN

Deficiency of ASM (acid sphingomyelinase) causes the lysosomal storage Niemann-Pick disease (NPD). Patients with NPD type B may develop progressive interstitial lung disease with frequent respiratory infections. Although several investigations using the ASM-deficient (ASMKO) mouse NPD model revealed inflammation and foamy macrophages, there is little insight into the pathogenesis of NPD-associated lung disease. Using ASMKO mice, we report that ASM deficiency is associated with a complex inflammatory phenotype characterized by marked accumulation of monocyte-derived CD11b+ macrophages and expansion of airspace/alveolar CD11c+ CD11b- macrophages, both with increased size, granularity, and foaminess. Both the alternative and classical pathways were activated, with decreased in situ phagocytosis of opsonized (Fc-coated) targets, preserved clearance of apoptotic cells (efferocytosis), secretion of Th2 cytokines, increased CD11c+/CD11b+ cells, and more than a twofold increase in lung and plasma proinflammatory cytokines. Macrophages, neutrophils, eosinophils, and noninflammatory lung cells of ASMKO lungs also exhibited marked accumulation of chitinase-like protein Ym1/2, which formed large eosinophilic polygonal Charcot-Leyden-like crystals. In addition to providing insight into novel features of lung inflammation that may be associated with NPD, our report provides a novel connection between ASM and the development of crystal-associated lung inflammation with alterations in macrophage biology.


Asunto(s)
Glicoproteínas/inmunología , Lisofosfolipasa/inmunología , Macrófagos Alveolares/inmunología , Macrófagos/inmunología , Enfermedad de Niemann-Pick Tipo A/inmunología , Enfermedad de Niemann-Pick Tipo B/inmunología , Neumonía/inmunología , Esfingomielina Fosfodiesterasa/inmunología , Animales , Antígenos CD11/genética , Antígenos CD11/inmunología , Antígeno CD11b/genética , Antígeno CD11b/inmunología , Tamaño de la Célula , Quitinasas/genética , Quitinasas/inmunología , Modelos Animales de Enfermedad , Eosinófilos/inmunología , Eosinófilos/patología , Femenino , Expresión Génica , Glicoproteínas/genética , Humanos , Lectinas/genética , Lectinas/inmunología , Pulmón/inmunología , Pulmón/patología , Lisofosfolipasa/genética , Macrófagos/patología , Macrófagos Alveolares/patología , Masculino , Ratones , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/patología , Enfermedad de Niemann-Pick Tipo A/enzimología , Enfermedad de Niemann-Pick Tipo A/genética , Enfermedad de Niemann-Pick Tipo A/patología , Enfermedad de Niemann-Pick Tipo B/enzimología , Enfermedad de Niemann-Pick Tipo B/genética , Enfermedad de Niemann-Pick Tipo B/patología , Fagocitosis , Neumonía/enzimología , Neumonía/genética , Neumonía/patología , Esfingomielina Fosfodiesterasa/deficiencia , Esfingomielina Fosfodiesterasa/genética , Balance Th1 - Th2/genética , beta-N-Acetilhexosaminidasas/genética , beta-N-Acetilhexosaminidasas/inmunología
6.
Arterioscler Thromb Vasc Biol ; 40(1): 61-71, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31619062

RESUMEN

OBJECTIVE: CD73 is an ectonucleotidase which catalyzes the conversion of AMP (adenosine monophosphate) to adenosine. Adenosine has been shown to be anti-inflammatory and vasorelaxant. The impact of ectonucleotidases on age-dependent atherosclerosis remains unclear. Our aim was to investigate the role of CD73 in age-dependent accumulation of atherosclerosis. Approach and results: Mice doubly deficient in CD73 and ApoE (apolipoprotein E; (cd73-/-/apoE-/-) were generated, and the extent of aortic atherosclerotic plaque was compared with apoE-/- controls at 12, 20, 32, and 52 weeks. By 12 weeks of age, cd73-/-/apoE-/- mice exhibited a significant increase in plaque (1.4±0.5% of the total vessel surface versus 0.4±0.1% in apoE-/- controls, P<0.005). By 20 weeks of age, this difference disappeared (2.9±0.4% versus 3.3±0.7%). A significant reversal in phenotype emerged at 32 weeks (9.8±1.2% versus 18.3±1.4%; P<0.0001) and persisted at the 52 week timepoint (22.4±2.1% versus 37.0±2.1%; P<0.0001). The inflammatory response to aging was found to be comparable between cd73-/-/apoE-/- mice and apoE-/- controls. A reduction in lipolysis in CD73 competent mice was observed, even with similar plasma lipid levels (cd73-/-/apoE-/- versus apoE-/- at 12 weeks [16.2±0.7 versus 9.5±1.4 nmol glycerol/well], 32 weeks [24.1±1.5 versus 7.4±0.4 nmol/well], and 52 weeks [13.8±0.62 versus 12.7±2.0 nmol/well], P<0.001). CONCLUSIONS: At early time points, CD73 exerts a subtle antiatherosclerotic influence, but with age, the pattern reverses, and the presence of CD73 promoted suppression of lipid catabolism.


Asunto(s)
5'-Nucleotidasa/genética , Aterosclerosis/genética , Regulación del Desarrollo de la Expresión Génica , ARN/genética , 5'-Nucleotidasa/biosíntesis , Animales , Aorta Torácica/metabolismo , Aorta Torácica/patología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo
8.
Am J Respir Cell Mol Biol ; 58(1): 66-78, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28850249

RESUMEN

Idiopathic pulmonary fibrosis is a progressive lung disease with complex pathophysiology and fatal prognosis. Macrophages (MΦ) contribute to the development of lung fibrosis; however, the underlying mechanisms and specific MΦ subsets involved remain unclear. During lung injury, two subsets of lung MΦ coexist: Siglec-Fhi resident alveolar MΦ and a mixed population of CD11bhi MΦ that primarily mature from immigrating monocytes. Using a novel inducible transgenic system driven by a fragment of the human CD68 promoter, we targeted deletion of the antiapoptotic protein cellular FADD-like IL-1ß-converting enzyme-inhibitory protein (c-FLIP) to CD11bhi MΦ. Upon loss of c-FLIP, CD11bhi MΦ became susceptible to cell death. Using this system, we were able to show that eliminating CD11bhi MΦ present 7-14 days after bleomycin injury was sufficient to protect mice from fibrosis. RNA-seq analysis of lung MΦ present during this time showed that CD11bhi MΦ, but not Siglec-Fhi MΦ, expressed high levels of profibrotic chemokines and growth factors. Human MΦ from patients with idiopathic pulmonary fibrosis expressed many of the same profibrotic chemokines identified in murine CD11bhi MΦ. Elimination of monocyte-derived MΦ may help in the treatment of fibrosis. We identify c-FLIP and the associated extrinsic cell death program as a potential pathway through which these profibrotic MΦ may be pharmacologically targeted.


Asunto(s)
Bleomicina/efectos adversos , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Antígenos CD11/metabolismo , Eliminación de Gen , Macrófagos/metabolismo , Fibrosis Pulmonar/metabolismo , Animales , Bleomicina/farmacología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Antígenos CD11/genética , Femenino , Humanos , Macrófagos/patología , Masculino , Ratones , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología
9.
J Immunol ; 196(3): 1366-75, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26718338

RESUMEN

Apoptotic cell (AC) clearance (efferocytosis) is an evolutionarily conserved process essential for immune health, particularly to maintain self-tolerance. Despite identification of many recognition receptors and intracellular signaling components of efferocytosis, its negative regulation remains incompletely understood and has not previously been known to involve microRNAs (miRs). In this article, we show that miR-34a (gene ID 407040), well recognized as a p53-dependent tumor suppressor, mediates coordinated negative regulation of efferocytosis by resident murine and human tissue macrophages (Mø). The miR-34a expression varied greatly between Mø from different tissues, correlating inversely with their capacity for AC uptake. Transient or genetic knockdown of miR-34a increased efferocytosis, whereas miR-34a overexpression decreased efferocytosis, without altering recognition of live, necrotic, or Ig-opsonized cells. The inhibitory effect of miR-34a was mediated both by reduced expression of Axl, a receptor tyrosine kinase known to recognize AC, and of the deacetylase silent information regulator T1, which had not previously been linked to efferocytosis by tissue Mø. Exposure to AC downregulated Mø miR-34a expression, resulting in a positive feedback loop that increased subsequent capacity to engulf AC. These findings demonstrate that miR-34a both specifically regulates and is regulated by efferocytosis. Given the ability of efferocytosis to polarize ingesting Mø uniquely and to reduce their host-defense functions, dynamic negative regulation by miR-34a provides one means of fine-tuning Mø behavior toward AC in specific tissue environments with differing potentials for microbial exposure.


Asunto(s)
Apoptosis/genética , Macrófagos/inmunología , MicroARNs/inmunología , Fagocitosis/genética , Sirtuina 1/inmunología , Animales , Apoptosis/inmunología , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Fagocitosis/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
10.
Am J Respir Cell Mol Biol ; 57(1): 66-76, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28257233

RESUMEN

The current paradigm in macrophage biology is that some tissues mainly contain macrophages from embryonic origin, such as microglia in the brain, whereas other tissues contain postnatal-derived macrophages, such as the gut. However, in the lung and in other organs, such as the skin, there are both embryonic and postnatal-derived macrophages. In this study, we demonstrate in the steady-state lung that the mononuclear phagocyte system is comprised of three newly identified interstitial macrophages (IMs), alveolar macrophages, dendritic cells, and few extravascular monocytes. We focused on similarities and differences between the three IM subtypes, specifically, their phenotype, location, transcriptional signature, phagocytic capacity, turnover, and lack of survival dependency on fractalkine receptor, CX3CR1. Pulmonary IMs were located in the bronchial interstitium but not the alveolar interstitium. At the transcriptional level, all three IMs displayed a macrophage signature and phenotype. All IMs expressed MER proto-oncogene, tyrosine kinase, CD64, CD11b, and CX3CR1, and were further distinguished by differences in cell surface protein expression of CD206, Lyve-1, CD11c, CCR2, and MHC class II, along with the absence of Ly6C, Ly6G, and Siglec F. Most intriguingly, in addition to the lung, similar phenotypic populations of IMs were observed in other nonlymphoid organs, perhaps highlighting conserved functions throughout the body. These findings promote future research to track four distinct pulmonary macrophages and decipher the division of labor that exists between them.


Asunto(s)
Pulmón/citología , Macrófagos/citología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Perfilación de la Expresión Génica , Macrófagos/metabolismo , Macrófagos Alveolares/citología , Macrófagos Alveolares/metabolismo , Ratones Endogámicos C57BL , Especificidad de Órganos , Fagocitos/citología , Fagocitos/metabolismo , Fenotipo , Transcripción Genética
11.
Am J Respir Cell Mol Biol ; 57(3): 294-306, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28421818

RESUMEN

Two populations of alveolar macrophages (AMs) coexist in the inflamed lung: resident AMs that arise during embryogenesis, and recruited AMs that originate postnatally from circulating monocytes. The objective of this study was to determine whether origin or environment dictates the transcriptional, metabolic, and functional programming of these two ontologically distinct populations over the time course of acute inflammation. RNA sequencing demonstrated marked transcriptional differences between resident and recruited AMs affecting three main areas: proliferation, inflammatory signaling, and metabolism. Functional assays and metabolomic studies confirmed these differences and demonstrated that resident AMs proliferate locally and are governed by increased tricarboxylic acid cycle and amino acid metabolism. Conversely, recruited AMs produce inflammatory cytokines in association with increased glycolytic and arginine metabolism. Collectively, the data show that even though they coexist in the same environment, inflammatory macrophage subsets have distinct immunometabolic programs and perform specialized functions during inflammation that are associated with their cellular origin.


Asunto(s)
Lesión Pulmonar Aguda/patología , Macrófagos/patología , Lesión Pulmonar Aguda/complicaciones , Lesión Pulmonar Aguda/genética , Animales , Linaje de la Célula , Proliferación Celular , Citocinas/metabolismo , Femenino , Perfilación de la Expresión Génica , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Masculino , Metabolómica , Ratones Endogámicos C57BL , Neumonía/complicaciones , Neumonía/genética , Neumonía/patología , Reproducibilidad de los Resultados , Análisis de Secuencia de ARN
12.
J Immunol ; 195(1): 174-84, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25987742

RESUMEN

Inhaled corticosteroids (ICS) increase community-acquired pneumonia (CAP) incidence in patients with chronic obstructive pulmonary disease (COPD) by unknown mechanisms. Apoptosis is increased in the lungs of COPD patients. Uptake of apoptotic cells (ACs) ("efferocytosis") by alveolar macrophages (AMøs) reduces their ability to combat microbes, including Streptococcus pneumoniae, the most common cause of CAP in COPD patients. Having shown that ICS significantly increase AMø efferocytosis, we hypothesized that this process, termed glucocorticoid-augmented efferocytosis, might explain the association of CAP with ICS therapy in COPD. To test this hypothesis, we studied the effects of fluticasone, AC, or both on AMøs of C57BL/6 mice in vitro and in an established model of pneumococcal pneumonia. Fluticasone plus AC significantly reduced TLR4-stimulated AMø IL-12 production, relative to either treatment alone, and decreased TNF-α, CCL3, CCL5, and keratinocyte-derived chemoattractant/CXCL1, relative to AC. Mice treated with fluticasone plus AC before infection with viable pneumococci developed significantly more lung CFUs at 48 h. However, none of the pretreatments altered inflammatory cell recruitment to the lungs at 48 h postinfection, and fluticasone plus AC less markedly reduced in vitro mediator production to heat-killed pneumococci. Fluticasone plus AC significantly reduced in vitro AMø killing of pneumococci, relative to other conditions, in part by delaying phagolysosome acidification without affecting production of reactive oxygen or nitrogen species. These results support glucocorticoid-augmented efferocytosis as a potential explanation for the epidemiological association of ICS therapy of COPD patients with increased risk for CAP, and establish murine experimental models to dissect underlying molecular mechanisms.


Asunto(s)
Corticoesteroides/efectos adversos , Androstadienos/efectos adversos , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Neumonía Neumocócica/inmunología , Animales , Apoptosis , Quimiocina CCL3/genética , Quimiocina CCL3/inmunología , Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Quimiocina CXCL1/genética , Quimiocina CXCL1/inmunología , Recuento de Colonia Microbiana , Modelos Animales de Enfermedad , Fluticasona , Regulación de la Expresión Génica , Humanos , Interleucina-12/genética , Interleucina-12/inmunología , Pulmón/microbiología , Pulmón/patología , Macrófagos Alveolares/microbiología , Macrófagos Alveolares/patología , Ratones , Ratones Endogámicos C57BL , Fagocitosis , Neumonía Neumocócica/inducido químicamente , Neumonía Neumocócica/genética , Neumonía Neumocócica/microbiología , Especies de Nitrógeno Reactivo/inmunología , Especies Reactivas de Oxígeno/inmunología , Streptococcus pneumoniae/inmunología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
14.
Am J Physiol Lung Cell Mol Physiol ; 311(1): L87-L100, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27190063

RESUMEN

During homeostasis two distinct macrophage (Mø) populations inhabit the lungs: tissue Mø (often called interstitial Mø) and resident alveolar Mø (resAMø). During acute lung inflammation, monocytes from the circulation migrate to areas of injury where they mature into a third Mø population: recruited Mø. Resident AMø uniquely express low levels of CD11b and high levels of CD11c. In comparison, recruited Mø and tissue Mø express high levels of CD11b and low levels of CD11c. It is likely that these three Mø subpopulations play distinct roles in injury and disease states; however, tools with which to individually target or track these populations are lacking. Here we demonstrate the utility of an hCD68-rtTA transgenic system for specific, robust, and inducible targeting of CD11b(+) recruited Mø and tissue Mø in the murine lung with negligible activation in resAMø. Using hCD68rtTA-GFP reporter mice, we show both during homeostasis and inflammation that administration of doxycycline induces tet-On reporter expression in recruited Mø and tissue Mø but not in resident AMø. We further demonstrate how hCD68-rtTA can be effectively combined with tet-On Cre to target these same recMø and tissue Mø. Accordingly, the hCD68-rtTA system is a powerful new tool that can be used for lineage tracing, fate mapping, and gene deletion in a variety of murine models, thereby enabling sophisticated investigation of the unique role of these CD11b(+) Mø during lung heath and disease.


Asunto(s)
Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/genética , Antígeno CD11b/metabolismo , Pulmón/patología , Fagocitos/metabolismo , Animales , Expresión Génica , Lipopolisacáridos/farmacología , Pulmón/inmunología , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos , Neumonía/metabolismo , Neumonía/patología , Activación Transcripcional
15.
J Immunol ; 189(1): 112-9, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22615206

RESUMEN

The lung environment actively inhibits apoptotic cell (AC) uptake by alveolar macrophages (AMøs) via lung collectin signaling through signal regulatory protein α (SIRPα). Even brief glucocorticoid (GC) treatment during maturation of human blood monocyte-derived or murine bone marrow-derived macrophages (Møs) increases their AC uptake. Whether GCs similarly impact differentiated tissue Møs and the mechanisms for this rapid response are unknown and important to define, given the widespread therapeutic use of inhaled GCs. We found that the GC fluticasone rapidly and dose-dependently increased AC uptake by murine AMøs without a requirement for protein synthesis. Fluticasone rapidly suppressed AMø expression of SIRPα mRNA and surface protein, and also activated a more delayed, translation-dependent upregulation of AC recognition receptors that was not required for the early increase in AC uptake. Consistent with a role for SIRPα suppression in rapid GC action, murine peritoneal Møs that had not been exposed to lung collectins showed delayed, but not rapid, increase in AC uptake. However, pretreatment of peritoneal Møs with the lung collectin surfactant protein D inhibited AC uptake, and fluticasone treatment rapidly reversed this inhibition. Thus, GCs act not only by upregulating AC recognition receptors during Mø maturation but also via a novel rapid downregulation of SIRPα expression by differentiated tissue Møs. Release of AMøs from inhibition of AC uptake by lung collectins may, in part, explain the beneficial role of inhaled GCs in inflammatory lung diseases, especially emphysema, in which there is both increased lung parenchymal cell apoptosis and defective AC uptake by AMøs.


Asunto(s)
Androstadienos/farmacología , Proteínas Reguladoras de la Apoptosis/farmacología , Apoptosis/inmunología , Colectinas/fisiología , Regulación hacia Abajo/inmunología , Tolerancia Inmunológica/inmunología , Macrófagos Alveolares/inmunología , Receptores Inmunológicos/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/fisiología , Células Cultivadas , Relación Dosis-Respuesta Inmunológica , Regulación hacia Abajo/efectos de los fármacos , Fluticasona , Tolerancia Inmunológica/efectos de los fármacos , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/patología , Ratones , Ratones Endogámicos C57BL , Receptores Inmunológicos/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
16.
Respir Res ; 14: 13, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23374856

RESUMEN

BACKGROUND: Toll-like receptors (TLRs) on T cells can modulate their responses, however, the extent and significance of TLR expression by lung T cells, NK cells, or NKT cells in chronic obstructive pulmonary disease (COPD) is unknown. METHODS: Lung tissue collected from clinically-indicated resections (n = 34) was used either: (a) to compare the expression of TLR1, TLR2, TLR2/1, TLR3, TLR4, TLR5, TLR6 and TLR9 on lung CD8+ T cells, CD4+ T cells, NK cells and NKT cells from smokers with or without COPD; or (b) to isolate CD8+ T cells for culture with anti-CD3ε without or with various TLR ligands. We measured protein expression of IFN-γ, TNF-α, IL-13, perforin, granzyme A, granzyme B, soluble FasL, CCL2, CCL3, CCL4, CCL5, CCL11, and CXCL9 in supernatants. RESULTS: All the lung subsets analyzed demonstrated low levels of specific TLR expression, but the percentage of CD8+ T cells expressing TLR1, TLR2, TLR4, TLR6 and TLR2/1 was significantly increased in COPD subjects relative to those without COPD. In contrast, from the same subjects, only TLR2/1 and TLR2 on lung CD4+ T cells and CD8+ NKT cells, respectively, showed a significant increase in COPD and there was no difference in TLR expression on lung CD56+ NK cells. Production of the Tc1 cytokines IFN-γ and TNF-α by lung CD8+ T cells were significantly increased via co-stimulation by Pam3CSK4, a specific TLR2/1 ligand, but not by other agonists. Furthermore, this increase in cytokine production was specific to lung CD8+ T cells from patients with COPD as compared to lung CD8+ T cells from smokers without COPD. CONCLUSIONS: These data suggest that as lung function worsens in COPD, the auto-aggressive behavior of lung CD8+ T cells could increase in response to microbial TLR ligands, specifically ligands against TLR2/1.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Regulación Bacteriana de la Expresión Génica , Pulmón/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Receptores Toll-Like/biosíntesis , Anciano , Linfocitos T CD8-positivos/microbiología , Células Cultivadas , Femenino , Humanos , Pulmón/citología , Pulmón/microbiología , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Enfermedad Pulmonar Obstructiva Crónica/microbiología
17.
Respir Res ; 14: 33, 2013 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-23497334

RESUMEN

BACKGROUND: Cigarette smoking is associated with increased frequency and duration of viral respiratory infections, but the underlying mechanisms are incompletely defined. We investigated whether smoking reduces expression by human lung macrophages (Mø) of receptors for viral nucleic acids and, if so, the effect on CXCL10 production. METHODS: We collected alveolar macrophages (AMø) by bronchoalveolar lavage of radiographically-normal lungs of subjects undergoing bronchoscopies for solitary nodules (n = 16) and of volunteers who were current or former smokers (n = 7) or never-smokers (n = 13). We measured expression of mRNA transcripts for viral nucleic acid receptors by real-time PCR in those AMø and in the human Mø cell line THP-1 following phorbol myristate acetate/vitamin D3 differentiation and exposure to cigarette smoke extract, and determined TLR3 protein expression using flow cytometry and immunohistochemistry. We also used flow cytometry to examine TLR3 expression in total lung Mø from subjects undergoing clinically-indicated lung resections (n = 25). Of these, seven had normal FEV1 and FEV1/FVC ratio (three former smokers, four current smokers); the remaining 18 subjects (14 former smokers; four current smokers) had COPD of GOLD stages I-IV. We measured AMø production of CXCL10 in response to stimulation with the dsRNA analogue poly(I:C) using Luminex assay. RESULTS: Relative to AMø of never-smokers, AMø of smokers demonstrated reduced protein expression of TLR3 and decreased mRNA for TLR3 but not TLR7, TLR8, TLR9, RIG-I, MDA-5 or PKR. Identical changes in TLR3 gene expression were induced in differentiated THP-1 cells exposed to cigarette smoke-extract in vitro for 4 hours. Among total lung Mø, the percentage of TLR3-positive cells correlated inversely with active smoking but not with COPD diagnosis, FEV1% predicted, sex, age or pack-years. Compared to AMø of never-smokers, poly(I:C)-stimulated production of CXCL10 was significantly reduced in AMø of smokers. CONCLUSIONS: Active smoking, independent of COPD stage or smoking duration, reduces both the percent of human lung Mø expressing TLR3, and dsRNA-induced CXCL10 production, without altering other endosomal or cytoplasmic receptors for microbial nucleic acids. This effect provides one possible mechanism for increased frequency and duration of viral lower respiratory tract infections in smokers. TRIAL REGISTRATION: ClinicalTrials.gov NCT00281190, NCT00281203 and NCT00281229.


Asunto(s)
Regulación hacia Abajo/genética , Macrófagos Alveolares/metabolismo , ARN Bicatenario/antagonistas & inhibidores , Fumar/metabolismo , Receptor Toll-Like 3/antagonistas & inhibidores , Adulto , Anciano , Línea Celular , Células Cultivadas , Estudios de Cohortes , Femenino , Humanos , Pulmón/citología , Pulmón/metabolismo , Pulmón/virología , Macrófagos Alveolares/virología , Masculino , Persona de Mediana Edad , ARN Bicatenario/genética , Fumar/genética , Receptor Toll-Like 3/biosíntesis , Receptor Toll-Like 3/genética , Adulto Joven
18.
Methods Mol Biol ; 2506: 257-267, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35771477

RESUMEN

Pulmonary macrophages are heterogeneous. Distinct populations of resident tissue macrophages exist in the lung airspace and tissue compartments during homeostasis. During inflammation, these are joined by monocyte-derived recruited macrophages. Flow cytometry can be used to identify and purify lung macrophage subsets. Here, we describe methods for identifying and isolating macrophages from bronchoalveolar lavage and digested lung tissues from mouse and human. We also describe basic staining for flow cytometry analysis of different macrophage subsets.


Asunto(s)
Pulmón , Macrófagos , Animales , Líquido del Lavado Bronquioalveolar , Citometría de Flujo/métodos , Humanos , Macrófagos Alveolares , Ratones
19.
Cell Rep ; 38(2): 110222, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35021097

RESUMEN

Phagocytosis of apoptotic cells, termed efferocytosis, is critical for tissue homeostasis and drives anti-inflammatory programming in engulfing macrophages. Here, we assess metabolites in naive and inflammatory macrophages following engulfment of multiple cellular and non-cellular targets. Efferocytosis leads to increases in the arginine-derived polyamines, spermidine and spermine, in vitro and in vivo. Surprisingly, polyamine accumulation after efferocytosis does not arise from retention of apoptotic cell metabolites or de novo synthesis but from enhanced polyamine import that is dependent on Rac1, actin, and PI3 kinase. Blocking polyamine import prevents efferocytosis from suppressing macrophage interleukin (IL)-1ß or IL-6. This identifies efferocytosis as a trigger for polyamine import and accumulation, and imported polyamines as mediators of efferocytosis-induced immune reprogramming.


Asunto(s)
Citofagocitosis/fisiología , Macrófagos/metabolismo , Poliaminas/metabolismo , Animales , Apoptosis/fisiología , Femenino , Voluntarios Sanos , Humanos , Inmunomodulación , Inflamación/metabolismo , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Espermidina/metabolismo , Espermina/metabolismo
20.
Methods Mol Biol ; 1809: 145-168, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29987790

RESUMEN

Transgenic mice have emerged as a central tool in the study of lung myeloid cells during homeostasis and disease. The use of Cre/Lox site-specific recombination allows for conditional deletion of a gene of interest in a spatially controlled manner. The basic Cre/Lox system can be further refined to include an inducible trigger, enabling conditional deletion of a gene of interest in a spatially and temporally controlled manner. Here we provide an overview of commercially available conditional and inducible conditional mouse strains that target lung myeloid cells and describe the appropriate breeding schemes and controls for transgenic animal systems that can be used to modulate myeloid cell function.


Asunto(s)
Células Mieloides/inmunología , Células Mieloides/metabolismo , Animales , Cruzamiento , Expresión Génica , Marcación de Gen , Genotipo , Técnicas de Genotipaje , Integrasas/genética , Integrasas/metabolismo , Mutación con Pérdida de Función , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Recombinación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA