Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
Add more filters

Publication year range
1.
Annu Rev Cell Dev Biol ; 33: 127-144, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28613937

ABSTRACT

In metazoans, removal of cells in situ is involved in larval maturation, metamorphosis, and embryonic development. In adults, such cell removal plays a role in the homeostatic maintenance of cell numbers and tissue integrity as well as in the response to cell injury and damage. This removal involves uptake of the whole or fragmented target cells into phagocytes. Depending on the organism, these latter may be near-neighbor tissue cells and/or professional phagocytes such as, in vertebrates, members of the myeloid family of cells, especially macrophages. The uptake processes appear to involve specialized and highly conserved recognition ligands and receptors, intracellular signaling in the phagocytes, and mechanisms for ingestion. The recognition of cells destined for this form of removal is critical and, significantly, is distinguished for the most part from the recognition of foreign materials and organisms by the innate and adaptive immune systems. In keeping with the key role of cell removal in maintaining tissue homeostasis, constant cell removal is normally silent, i.e., does not initiate a local tissue reaction. This article discusses these complex and wide-ranging processes in general terms as well as the implications when these processes are disrupted in inflammation, immunity, and disease.


Subject(s)
Phagocytosis , Animals , Apoptosis , Disease , Homeostasis , Humans , Intracellular Space/metabolism , Phagocytes/cytology
2.
Blood ; 139(11): 1707-1721, 2022 03 17.
Article in English | MEDLINE | ID: mdl-34699591

ABSTRACT

Loss of NADPH oxidase activity leads to altered phagocyte responses and exaggerated inflammation in chronic granulomatous disease (CGD). We sought to assess the effects of Nox2 absence on monocyte-derived macrophages (MoMacs) in gp91phox-/y mice during zymosan-induced peritonitis. MoMacs from CGD and wild-type (WT) peritonea were characterized over time after zymosan injection. Although numbers lavaged from both genotypes were virtually identical, there were marked differences in maturation: newly recruited WT MoMacs rapidly enlarged and matured, losing Ly6C and gaining MHCII, CD206, and CD36, whereas CGD MoMacs remained small and were mostly Ly6C+MHCII-. RNA-sequencing analyses showed few intrinsic differences between genotypes in newly recruited MoMacs but significant differences with time. WT MoMacs displayed changes in metabolism, adhesion, and reparative functions, whereas CGD MoMacs remained inflammatory. PKH dye labeling revealed that although WT MoMacs were mostly recruited within the first 24 hours and remained in the peritoneum while maturing and enlarging, CGD monocytes streamed into the peritoneum for days, with many migrating to the diaphragm where they were found in fibrin(ogen) clots surrounding clusters of neutrophils in nascent pyogranulomata. Importantly, these observations seemed to be driven by milieu: adoptive transfer of CGD MoMacs into inflamed peritonea of WT mice resulted in immunophenotypic maturation and normal behavior, whereas altered maturation/behavior of WT MoMacs resulted from transfer into inflamed peritonea of CGD mice. In addition, Nox2-deficient MoMacs behaved similarly to their Nox2-sufficient counterparts within the largely WT milieu of mixed bone marrow chimeras. These data show persistent recruitment with fundamental failure of MoMac maturation in CGD.


Subject(s)
Granulomatous Disease, Chronic , Animals , Granulomatous Disease, Chronic/genetics , Inflammation/metabolism , Macrophages/metabolism , Mice , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Neutrophils/metabolism
3.
Am J Physiol Lung Cell Mol Physiol ; 324(4): L536-L549, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36852927

ABSTRACT

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.


Subject(s)
Folate Receptor 2 , Pneumonia , Humans , Monocytes/metabolism , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Macrophages/metabolism , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/metabolism , Inflammation/genetics , Inflammation/metabolism , Sequence Analysis, RNA/methods , Folate Receptor 2/metabolism
4.
Am J Physiol Lung Cell Mol Physiol ; 323(4): L391-L399, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35943156

ABSTRACT

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.


Subject(s)
Cigarette Smoking , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Animals , Disease Models, Animal , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Leukocytes , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Emphysema/pathology , Nicotiana/adverse effects , X-Ray Microtomography
5.
Immunity ; 39(3): 599-610, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-24012416

ABSTRACT

It is thought that monocytes rapidly differentiate to macrophages or dendritic cells (DCs) upon leaving blood. Here we have shown that Ly-6C⁺ monocytes constitutively trafficked into skin, lung, and lymph nodes (LNs). Entry was unaffected in gnotobiotic mice. Monocytes in resting lung and LN had similar gene expression profiles to blood monocytes but elevated transcripts of a limited number of genes including cyclo-oxygenase-2 (COX-2) and major histocompatibility complex class II (MHCII), induced by monocyte interaction with endothelium. Parabiosis, bromodoxyuridine (BrdU) pulse-chase analysis, and intranasal instillation of tracers indicated that instead of contributing to resident macrophages in the lung, recruited endogenous monocytes acquired antigen for carriage to draining LNs, a function redundant with DCs though differentiation to DCs did not occur. Thus, monocytes can enter steady-state nonlymphoid organs and recirculate to LNs without differentiation to macrophages or DCs, revising a long-held view that monocytes become tissue-resident macrophages by default.


Subject(s)
Cell Differentiation , Dendritic Cells/metabolism , Lymph Nodes/cytology , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Animals , Antigens, Ly/metabolism , Cell Movement , Cyclooxygenase 2/genetics , Dendritic Cells/cytology , Dendritic Cells/immunology , Endothelium/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Lung/cytology , Lymph Nodes/immunology , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Skin/cytology
6.
Am J Respir Crit Care Med ; 203(8): 946-956, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33079572

ABSTRACT

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.


Subject(s)
Bronchoalveolar Lavage Fluid/immunology , Gene Expression Profiling , Leukocytes, Mononuclear/immunology , Macrophages, Alveolar/immunology , Monocytes/immunology , Pulmonary Alveoli/immunology , Sequence Analysis, RNA , Adult , Female , Healthy Volunteers , Humans , Male , Middle Aged , Sex Factors
7.
Immunity ; 36(5): 807-20, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22542147

ABSTRACT

Toll-like receptor (TLR) stimulation activates macrophages to resist intracellular pathogens. Yet, the intracellular bacterium Listeria monocytogenes (Lm) causes lethal infections in spite of innate immune cell activation. Lm uses direct cell-cell spread to disseminate within its host. Here, we have shown that TLR-activated macrophages killed cell-free Lm but failed to prevent infection by spreading Lm. Instead, TLR signals increased the efficiency of Lm spread from "donor" to "recipient" macrophages. This enhancement required nitric oxide (NO) production by nitric oxide synthase-2 (NOS2). NO increased Lm escape from secondary vacuoles in recipient cells and delayed maturation of phagosomes containing membrane-like particles that mimic Lm-containing pseudopods. NO also promoted Lm spread during systemic in vivo infection, as shown by the fact that inhibition of NOS2 with 1400W reduced spread-dependent Lm burdens in mouse livers. These findings reveal a mechanism by which pathogens capable of cell-cell spread can avoid the consequences of innate immune cell activation by TLR stimuli.


Subject(s)
Listeria monocytogenes/immunology , Listeriosis/immunology , Macrophages/immunology , Macrophages/metabolism , Nitric Oxide/metabolism , Toll-Like Receptors/metabolism , Animals , Cells, Cultured , Immunity, Innate/immunology , Listeria monocytogenes/metabolism , Listeriosis/metabolism , Macrophage Activation/immunology , Mice , Mice, Inbred C57BL , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/immunology , Nitric Oxide Synthase Type II/immunology , Nitric Oxide Synthase Type II/metabolism , Phagosomes/immunology , Phagosomes/metabolism , Toll-Like Receptors/immunology
8.
Am J Respir Crit Care Med ; 201(10): 1209-1217, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32197050

ABSTRACT

Rationale: Interstitial macrophages (IMs) and airspace macrophages (AMs) play critical roles in lung homeostasis and host defense, and are central to the pathogenesis of a number of lung diseases. However, the absolute numbers of macrophages and the precise anatomic locations they occupy in the healthy human lung have not been quantified.Objectives: To determine the precise number and anatomic location of human pulmonary macrophages in nondiseased lungs and to quantify how this is altered in chronic cigarette smokers.Methods: Whole right upper lobes from 12 human donors without pulmonary disease (6 smokers and 6 nonsmokers) were evaluated using design-based stereology. CD206 (cluster of differentiation 206)-positive/CD43+ AMs and CD206+/CD43- IMs were counted in five distinct anatomical locations using the optical disector probe.Measurements and Main Results: An average of 2.1 × 109 IMs and 1.4 × 109 AMs were estimated per right upper lobe. Of the AMs, 95% were contained in diffusing airspaces and 5% in airways. Of the IMs, 78% were located within the alveolar septa, 14% around small vessels, and 7% around the airways. The local density of IMs was greater in the alveolar septa than in the connective tissue surrounding the airways or vessels. The total number and density of IMs was 36% to 56% greater in the lungs of cigarette smokers versus nonsmokers.Conclusions: The precise locations occupied by pulmonary macrophages were defined in nondiseased human lungs from smokers and nonsmokers. IM density was greatest in the alveolar septa. Lungs from chronic smokers had increased IM numbers and overall density, supporting a role for IMs in smoking-related disease.


Subject(s)
Cigarette Smoking/pathology , Lung/pathology , Macrophages, Alveolar/pathology , Adolescent , Adult , Aged , Case-Control Studies , Cell Count , Female , Humans , Immunohistochemistry , Lectins, C-Type/metabolism , Leukosialin/metabolism , Lung/cytology , Lung/metabolism , Macrophages, Alveolar/metabolism , Male , Mannose Receptor , Mannose-Binding Lectins/metabolism , Middle Aged , Optical Devices , Receptors, Cell Surface/metabolism , Tissue Donors
9.
Nature ; 505(7483): 412-6, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24317696

ABSTRACT

Respiratory surfaces are exposed to billions of particulates and pathogens daily. A protective mucus barrier traps and eliminates them through mucociliary clearance (MCC). However, excessive mucus contributes to transient respiratory infections and to the pathogenesis of numerous respiratory diseases. MUC5AC and MUC5B are evolutionarily conserved genes that encode structurally related mucin glycoproteins, the principal macromolecules in airway mucus. Genetic variants are linked to diverse lung diseases, but specific roles for MUC5AC and MUC5B in MCC, and the lasting effects of their inhibition, are unknown. Here we show that mouse Muc5b (but not Muc5ac) is required for MCC, for controlling infections in the airways and middle ear, and for maintaining immune homeostasis in mouse lungs, whereas Muc5ac is dispensable. Muc5b deficiency caused materials to accumulate in upper and lower airways. This defect led to chronic infection by multiple bacterial species, including Staphylococcus aureus, and to inflammation that failed to resolve normally. Apoptotic macrophages accumulated, phagocytosis was impaired, and interleukin-23 (IL-23) production was reduced in Muc5b(-/-) mice. By contrast, in mice that transgenically overexpress Muc5b, macrophage functions improved. Existing dogma defines mucous phenotypes in asthma and chronic obstructive pulmonary disease (COPD) as driven by increased MUC5AC, with MUC5B levels either unaffected or increased in expectorated sputum. However, in many patients, MUC5B production at airway surfaces decreases by as much as 90%. By distinguishing a specific role for Muc5b in MCC, and by determining its impact on bacterial infections and inflammation in mice, our results provide a refined framework for designing targeted therapies to control mucin secretion and restore MCC.


Subject(s)
Lung/immunology , Mucin-5B/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Animals , Asthma/immunology , Asthma/metabolism , Bacterial Infections/immunology , Bacterial Infections/microbiology , Cilia/physiology , Ear, Middle/immunology , Ear, Middle/microbiology , Female , Inflammation/pathology , Lung/metabolism , Lung/microbiology , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Mucin 5AC/deficiency , Mucin 5AC/metabolism , Mucin-5B/deficiency , Mucin-5B/genetics , Phagocytosis , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/microbiology , Staphylococcus aureus/immunology , Survival Analysis
10.
Am J Respir Cell Mol Biol ; 59(5): 580-591, 2018 11.
Article in English | MEDLINE | ID: mdl-29953261

ABSTRACT

Early recognition of neoantigen-expressing cells is complex, involving multiple immune cell types. In this study, in vivo, we examined how antigen-presenting cell subtypes coordinate and induce an immunological response against neoantigen-expressing cells, particularly in the absence of a pathogen-associated molecular pattern, which is normally required to license antigen-presenting cells to present foreign or self-antigens as immunogens. Using two reductionist models of neoantigen-expressing cells and two cancer models, we demonstrated that natural IgM is essential for the recognition and initiation of adaptive immunity against neoantigen-expressing cells. Natural IgM antibodies form a cellular immune complex with the neoantigen-expressing cells. This immune complex licenses surveying monocytes to present neoantigens as immunogens to CD4+ T cells. CD4+ T helper cells, in turn, use CD40L to license cross-presenting CD40+ Batf3+ dendritic cells to elicit a cytotoxic T cell response against neoantigen-expressing cells. Any break along this immunological chain reaction results in the escape of neoantigen-expressing cells. This study demonstrates the surprising, essential role of natural IgM as the initiator of a sequential signaling cascade involving multiple immune cell subtypes. This sequence is required to coordinate an adaptive immune response against neoantigen-expressing cells.


Subject(s)
Adaptive Immunity , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunoglobulin M/immunology , Lung Neoplasms/immunology , Melanoma, Experimental/immunology , Animals , Antigen-Presenting Cells/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/pathology , Female , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lymphocyte Activation , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monocytes/immunology , T-Lymphocytes, Helper-Inducer/immunology
11.
Am J Respir Cell Mol Biol ; 58(1): 66-78, 2018 01.
Article in English | MEDLINE | ID: mdl-28850249

ABSTRACT

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.


Subject(s)
Bleomycin/adverse effects , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , CD11 Antigens/metabolism , Gene Deletion , Macrophages/metabolism , Pulmonary Fibrosis/metabolism , Animals , Bleomycin/pharmacology , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CD11 Antigens/genetics , Female , Humans , Macrophages/pathology , Male , Mice , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology
12.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L69-L82, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28935638

ABSTRACT

Microparticles are a newly recognized class of mediators in the pathophysiology of lung inflammation and injury, but little is known about the factors that regulate their accumulation and clearance. The primary objective of our study was to determine whether alveolar macrophages engulf microparticles and to elucidate the mechanisms by which this occurs. Alveolar microparticles were quantified in bronchoalveolar fluid of mice with lung injury induced by LPS and hydrochloric acid. Microparticle numbers were greatest at the peak of inflammation and declined as inflammation resolved. Isolated, fluorescently labeled particles were placed in culture with macrophages to evaluate ingestion in the presence of endocytosis inhibitors. Ingestion was blocked with cytochalasin D and wortmannin, consistent with a phagocytic process. In separate experiments, mice were treated intratracheally with labeled microparticles, and their uptake was assessed though microscopy and flow cytometry. Resident alveolar macrophages, not recruited macrophages, were the primary cell-ingesting microparticles in the alveolus during lung injury. In vitro, microparticles promoted inflammatory signaling in LPS primed epithelial cells, signifying the importance of microparticle clearance in resolving lung injury. Microparticles were found to have phosphatidylserine exposed on their surfaces. Accordingly, we measured expression of phosphatidylserine receptors on macrophages and found high expression of MerTK and Axl in the resident macrophage population. Endocytosis of microparticles was markedly reduced in MerTK-deficient macrophages in vitro and in vivo. In conclusion, microparticles are released during acute lung injury and peak in number at the height of inflammation. Resident alveolar macrophages efficiently clear these microparticles through MerTK-mediated phagocytosis.


Subject(s)
Acute Lung Injury/physiopathology , Cell-Derived Microparticles/physiology , Inflammation/pathology , Macrophages, Alveolar/physiology , Phagocytosis , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , c-Mer Tyrosine Kinase/physiology , Acute Lung Injury/metabolism , Animals , Apoptosis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , Axl Receptor Tyrosine Kinase
13.
J Immunol ; 197(4): 1425-34, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27402702

ABSTRACT

Proinflammatory consequences have been described for lysophosphatidylcholine, a lipid product of cellular injury, signaling via the G protein-coupled receptor G2A on myeloid and lymphoid inflammatory cells. This prompted the hypothesis that genetic deletion of G2A would limit intestinal inflammation in a mouse model of colitis induced by dextran sodium sulfate. Surprisingly, G2A(-/-) mice exhibited significantly worsened colitis compared with wild-type mice, as demonstrated by disease activity, colon shortening, histology, and elevated IL-6 and IL-5 in colon tissues. Investigation of inflammatory cells recruited to inflamed G2A(-/-) colons showed significantly more TNF-α(+) and Ly6C(hi)MHCII(-) proinflammatory monocytes and eosinophils than in wild-type colons. Both monocytes and eosinophils were pathogenic as their depletion abolished the excess inflammation in G2A(-/-) mice. G2A(-/-) mice also had less IFN-γ in inflamed colon tissues than wild-type mice. Fewer CD4(+) lymphocytes were recruited to inflamed G2A(-/-) colons, and fewer colonic lymphocytes produced IFN-γ upon ex vivo stimulation. Administration of IFN-γ to G2A(-/-) mice during dextran sodium sulfate exposure abolished the excess colitic inflammation and reduced colonic IL-5 and eosinophil numbers to levels seen in wild-type mice. Furthermore, IFN-γ reduced the numbers of TNF-α(+) monocyte and enhanced their maturation from Ly6C(hi)MHCII(-) to Ly6C(int)MHCII(+) Taken together, the data suggest that G2A signaling serves to dampen intestinal inflammation via the production of IFN-γ, which, in turn, enhances monocyte maturation to a less inflammatory program and ultimately reduces eosinophil-induced injury of colonic tissues.


Subject(s)
Cell Cycle Proteins/metabolism , Colitis/pathology , Interferon-gamma/biosynthesis , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Animals , Colitis/metabolism , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Flow Cytometry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
14.
Am J Respir Cell Mol Biol ; 57(5): 519-526, 2017 11.
Article in English | MEDLINE | ID: mdl-28586241

ABSTRACT

The alveolar epithelium consists of squamous alveolar type (AT) I and cuboidal ATII cells. ATI cells cover 95-98% of the alveolar surface, thereby playing a critical role in barrier integrity, and are extremely thin, thus permitting efficient gas exchange. During lung injury, ATI cells die, resulting in increased epithelial permeability. ATII cells re-epithelialize the alveolar surface via proliferation and transdifferentiation into ATI cells. Transdifferentiation is characterized by down-regulation of ATII cell markers, up-regulation of ATI cell markers, and cell spreading, resulting in a change in morphology from cuboidal to squamous, thus restoring normal alveolar architecture and function. The mechanisms underlying ATII to ATI cell transdifferentiation have not been well studied in vivo. A prerequisite for mechanistic investigation is a rigorous, unbiased method to quantitate this process. Here, we used SPCCreERT2;mTmG mice, in which ATII cells and their progeny express green fluorescent protein (GFP), and applied stereologic techniques to measure transdifferentiation during repair after injury induced by LPS. Transdifferentiation was quantitated as the percent of alveolar surface area covered by ATII-derived (GFP+) cells expressing ATI, but not ATII, cell markers. Using this methodology, the time course and magnitude of transdifferentiation during repair was determined. We found that ATI cell loss and epithelial permeability occurred by Day 4, and ATII to ATI cell transdifferentiation began by Day 7 and continued until Day 16. Notably, transdifferentiation and barrier restoration are temporally correlated. This methodology can be applied to investigate the molecular mechanisms underlying transdifferentiation, ultimately revealing novel therapeutic targets to accelerate repair after lung injury.


Subject(s)
Alveolar Epithelial Cells/pathology , Cell Transdifferentiation/physiology , Lung Injury/pathology , Pulmonary Alveoli/pathology , Animals , Cell Proliferation/physiology , Cells, Cultured , Epithelium/pathology , Mice, Transgenic
15.
Am J Respir Cell Mol Biol ; 57(1): 66-76, 2017 07.
Article in English | MEDLINE | ID: mdl-28257233

ABSTRACT

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.


Subject(s)
Lung/cytology , Macrophages/cytology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Gene Expression Profiling , Macrophages/metabolism , Macrophages, Alveolar/cytology , Macrophages, Alveolar/metabolism , Mice, Inbred C57BL , Organ Specificity , Phagocytes/cytology , Phagocytes/metabolism , Phenotype , Transcription, Genetic
16.
J Immunol ; 195(1): 46-50, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26034174

ABSTRACT

In transplantation, a major obstacle for graft acceptance in MHC-matched individuals is the mismatch of minor histocompatibility Ags. Minor histocompatibility Ags are peptides derived from polymorphic proteins that can be presented by APCs on MHC molecules. The APC subtype uniquely responsible for the rejection of minor Ag-mismatched grafts has not yet been identified. In this study, we examined graft rejection in three mouse models: 1) mismatch of male-specific minor Ags, 2) mismatch of minor Ags distinct from male-specific minor Ags, and 3) skin transplant. This study demonstrates that in the absence of pathogen-associated molecular patterns, Batf3-dependent dendritic cells elicit the rejection of cells and grafts expressing mismatched minor Ags. The implication of our findings in clinical transplantation may be significant, as minor Ag reactivity has been implicated in the pathogenesis of multiple allograft tissues.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Dendritic Cells/immunology , Gene Expression Regulation, Developmental , Graft Rejection , Minor Histocompatibility Antigens/immunology , Repressor Proteins/immunology , Skin Transplantation , Adoptive Transfer , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Dendritic Cells/cytology , Female , Histocompatibility Testing , Lymph Nodes/cytology , Lymph Nodes/immunology , Male , Mice , Mice, Knockout , Minor Histocompatibility Antigens/genetics , Repressor Proteins/genetics , Signal Transduction , Spleen/cytology , Spleen/immunology , Transplantation, Homologous
17.
Am J Respir Crit Care Med ; 193(6): 614-26, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26551758

ABSTRACT

RATIONALE: The pulmonary mononuclear phagocyte system is a critical host defense mechanism composed of macrophages, monocytes, monocyte-derived cells, and dendritic cells. However, our current characterization of these cells is limited because it is derived largely from animal studies and analysis of human mononuclear phagocytes from blood and small tissue resections around tumors. OBJECTIVES: Phenotypic and morphologic characterization of mononuclear phagocytes that potentially access inhaled antigens in human lungs. METHODS: We acquired and analyzed pulmonary mononuclear phagocytes from fully intact nondiseased human lungs (including the major blood vessels and draining lymph nodes) obtained en bloc from 72 individual donors. Differential labeling of hematopoietic cells via intrabronchial and intravenous administration of antibodies within the same lobe was used to identify extravascular tissue-resident mononuclear phagocytes and exclude cells within the vascular lumen. Multiparameter flow cytometry was used to identify mononuclear phagocyte populations among cells labeled by each route of antibody delivery. MEASUREMENTS AND MAIN RESULTS: We performed a phenotypic analysis of pulmonary mononuclear phagocytes isolated from whole nondiseased human lungs and lung-draining lymph nodes. Five pulmonary mononuclear phagocytes were observed, including macrophages, monocyte-derived cells, and dendritic cells that were phenotypically distinct from cell populations found in blood. CONCLUSIONS: Different mononuclear phagocytes, particularly dendritic cells, were labeled by intravascular and intrabronchial antibody delivery, countering the notion that tissue and blood mononuclear phagocytes are equivalent systems. Phenotypic descriptions of the mononuclear phagocytes in nondiseased lungs provide a precedent for comparative studies in diseased lungs and potential targets for therapeutics.


Subject(s)
Flow Cytometry , Lung/immunology , Lymph Nodes/immunology , Mononuclear Phagocyte System/immunology , Phagocytes/immunology , Adult , Cadaver , Female , Humans , Male
18.
J Allergy Clin Immunol ; 135(2): 517-527.e12, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25498313

ABSTRACT

BACKGROUND: Deficient production of reactive oxygen species (ROS) by the phagocyte nicotinamide adenine dinucleotide (NADPH) oxidase in patients with chronic granulomatous disease (CGD) results in susceptibility to certain pathogens secondary to impaired oxidative killing and mobilization of other phagocyte defenses. Peroxisome proliferator-activated receptor (PPAR) γ agonists, including pioglitazone, approved for type 2 diabetes therapy alter cellular metabolism and can heighten ROS production. It was hypothesized that pioglitazone treatment of gp91(phox-/-) mice, a murine model of human CGD, would enhance phagocyte oxidant production and killing of Staphylococcus aureus, a significant pathogen in patients with this disorder. OBJECTIVES: We sought to determine whether pioglitazone treatment of gp91(phox-/-) mice enhanced phagocyte oxidant production and host defense. METHODS: Wild-type and gp91(phox-/-) mice were treated with the PPARγ agonist pioglitazone, and phagocyte ROS and killing of S aureus were investigated. RESULTS: As demonstrated by 3 different ROS-sensing probes, short-term treatment of gp91(phox-/-) mice with pioglitazone enhanced stimulated ROS production in neutrophils and monocytes from blood and neutrophils and inflammatory macrophages recruited to tissues. Mitochondria were identified as the source of ROS. Findings were replicated in human monocytes from patients with CGD after ex vivo pioglitazone treatment. Importantly, although mitochondrial (mt)ROS were deficient in gp91(phox-/-) phagocytes, their restoration with treatment significantly enabled killing of S aureus both ex vivo and in vivo. CONCLUSIONS: Together, the data support the hypothesis that signaling from the NADPH oxidase under normal circumstances governs phagocyte mtROS production and that such signaling is lacking in the absence of a functioning phagocyte oxidase. PPARγ agonism appears to bypass the need for the NADPH oxidase for enhanced mtROS production and partially restores host defense in CGD.


Subject(s)
Granulomatous Disease, Chronic/immunology , Granulomatous Disease, Chronic/metabolism , Mitochondria/metabolism , Oxidants/metabolism , Phagocytes/immunology , Phagocytes/metabolism , Thiazolidinediones/pharmacology , Animals , Disease Models, Animal , Humans , Male , Membrane Glycoproteins/deficiency , Mice , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , NADPH Oxidase 2 , NADPH Oxidases/deficiency , Neutrophils/immunology , Neutrophils/metabolism , PPAR gamma/metabolism , Phagocytes/microbiology , Phagocytosis/immunology , Pioglitazone , Reactive Oxygen Species/metabolism , Staphylococcus aureus/immunology , Superoxides/metabolism
19.
Am J Pathol ; 184(5): 1489-502, 2014 May.
Article in English | MEDLINE | ID: mdl-24650563

ABSTRACT

Fibrotic lung diseases represent a diverse group of progressive and often fatal disorders with limited treatment options. Although the pathogenesis of these conditions remains incompletely understood, receptor type protein tyrosine phosphatase α (PTP-α encoded by PTPRA) has emerged as a key regulator of fibroblast signaling. We previously reported that PTP-α regulates cellular responses to cytokines and growth factors through integrin-mediated signaling and that PTP-α promotes fibroblast expression of matrix metalloproteinase 3, a matrix-degrading proteinase linked to pulmonary fibrosis. Here, we sought to determine more directly the role of PTP-α in pulmonary fibrosis. Mice genetically deficient in PTP-α (Ptpra(-/-)) were protected from pulmonary fibrosis induced by intratracheal bleomycin, with minimal alterations in the early inflammatory response or production of TGF-ß. Ptpra(-/-) mice were also protected from pulmonary fibrosis induced by adenoviral-mediated expression of active TGF-ß1. In reciprocal bone marrow chimera experiments, the protective phenotype tracked with lung parenchymal cells but not bone marrow-derived cells. Because fibroblasts are key contributors to tissue fibrosis, we compared profibrotic responses in wild-type and Ptpra(-/-) mouse embryonic and lung fibroblasts. Ptpra(-/-) fibroblasts exhibited hyporesponsiveness to TGF-ß, manifested by diminished expression of αSMA, EDA-fibronectin, collagen 1A, and CTGF. Ptpra(-/-) fibroblasts exhibited markedly attenuated TGF-ß-induced Smad2/3 transcriptional activity. We conclude that PTP-α promotes profibrotic signaling pathways in fibroblasts through control of cellular responsiveness to TGF-ß.


Subject(s)
Fibroblasts/pathology , Lung/pathology , Pulmonary Fibrosis/pathology , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Adenoviridae , Animals , Bleomycin , Cytokines/biosynthesis , Gene Deletion , Genes, Reporter , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Pneumonia/complications , Pneumonia/pathology , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/prevention & control , Receptor-Like Protein Tyrosine Phosphatases, Class 4/deficiency , Receptors, Transforming Growth Factor beta/metabolism , Smad Proteins/metabolism , Transcription, Genetic
20.
Am J Respir Cell Mol Biol ; 50(4): 825-37, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24325577

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a relentless, fibrotic parenchymal lung disease in which alternatively programmed macrophages produce profibrotic molecules that promote myofibroblast survival and collagen synthesis. Effective therapies to treat patients with IPF are lacking, and conventional therapy may be harmful. We tested the hypothesis that therapeutic lung delivery of the proinflammatory cytokine tumor necrosis factor (TNF)-α into wild-type fibrotic mice would reduce the profibrotic milieu and accelerate the resolution of established pulmonary fibrosis. Fibrosis was assessed in bleomycin-instilled wild-type and TNF-α(-/-) mice by measuring hydroxyproline levels, static compliance, and Masson's trichrome staining. Macrophage infiltration and programming status was assessed by flow cytometry of enzymatically digested lung and in situ immunostaining. Pulmonary delivery of TNF-α to wild-type mice with established pulmonary fibrosis was found to reduce their fibrotic burden, to improve lung function and architecture, and to reduce the number and programming status of profibrotic alternatively programmed macrophages. In contrast, fibrosis and alternative macrophage programming were prolonged in bleomycin-instilled TNF-α(-/-) mice. To address the role of the reduced numbers of alternatively programmed macrophages in the TNF-α-induced resolution of established pulmonary fibrosis, we conditionally depleted macrophages in MAFIA (MAcrophage Fas-Induced Apoptosis) mice. Conditional macrophage depletion phenocopied the resolution of established pulmonary fibrosis observed after therapeutic TNF-α delivery. Taken together, our results show for the first time that TNF-α is involved in the resolution of established pulmonary fibrosis via a mechanism involving reduced numbers and programming status of profibrotic macrophages. We speculate that pulmonary delivery of TNF-α or augmenting its signaling pathway represent a novel therapeutic strategy to resolve established pulmonary fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis/drug therapy , Lung/drug effects , Macrophages, Alveolar/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Bleomycin , Cells, Cultured , Disease Models, Animal , Hydroxyproline/metabolism , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/physiopathology , Lung/metabolism , Lung/pathology , Lung/physiopathology , Lung Compliance , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptor, Macrophage Colony-Stimulating Factor/genetics , Recovery of Function , Remission Induction , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Time Factors , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics , fas Receptor/genetics , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL