Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 40
1.
Elife ; 102021 09 20.
Article En | MEDLINE | ID: mdl-34544549

Vaccination strategies for rapid protection against multidrug-resistant bacterial infection are very important, especially for hospitalized patients who have high risk of exposure to these bacteria. However, few such vaccination strategies exist due to a shortage of knowledge supporting their rapid effect. Here, we demonstrated that a single intranasal immunization of inactivated whole cell of Acinetobacter baumannii elicits rapid protection against broad A. baumannii-infected pneumonia via training of innate immune response in Rag1-/- mice. Immunization-trained alveolar macrophages (AMs) showed enhanced TNF-α production upon restimulation. Adoptive transfer of immunization-trained AMs into naive mice mediated rapid protection against infection. Elevated TLR4 expression on vaccination-trained AMs contributed to rapid protection. Moreover, immunization-induced rapid protection was also seen in Pseudomonas aeruginosa and Klebsiella pneumoniae pneumonia models, but not in Staphylococcus aureus and Streptococcus pneumoniae model. Our data reveal that a single intranasal immunization induces rapid and efficient protection against certain Gram-negative bacterial pneumonia via training AMs response, which highlights the importance and the possibility of harnessing trained immunity of AMs to design rapid-effecting vaccine.


Acinetobacter Infections/prevention & control , Acinetobacter baumannii/immunology , Bacterial Vaccines/administration & dosage , Klebsiella Infections/prevention & control , Klebsiella pneumoniae/immunology , Macrophages, Alveolar/drug effects , Pneumonia, Bacterial/prevention & control , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/immunology , Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Administration, Intranasal , Adoptive Transfer , Animals , Cells, Cultured , Disease Models, Animal , Female , Homeodomain Proteins/genetics , Immunity, Innate/drug effects , Klebsiella Infections/immunology , Klebsiella Infections/microbiology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/transplantation , Mice, Inbred C57BL , Mice, Knockout , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Time Factors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vaccination , Vaccines, Inactivated/administration & dosage
2.
Am J Physiol Lung Cell Mol Physiol ; 321(4): L787-L801, 2021 10 01.
Article En | MEDLINE | ID: mdl-34405715

Mechanical ventilation is a life-sustaining therapy for patients with respiratory failure but can cause further lung damage known as ventilator-induced lung injury (VILI). However, the intrinsic molecular mechanisms underlying recovery of VILI remain unknown. Phagocytosis of apoptotic cells (also known as efferocytosis) is a key mechanism orchestrating successful resolution of inflammation. Here we show the positive regulation of macrophage Toll-like receptor (TLR) 4 in efferocytosis and resolution of VILI. Mice were depleted of alveolar macrophages and then subjected to injurious ventilation (tidal volume, 20 mL/kg) for 4 h. On day 1 after mechanical ventilation, Tlr4+/+ or Tlr4-/- bone marrow-derived macrophages (BMDMs) were intratracheally administered to alveolar macrophage-depleted mice. We observed that mice depleted of alveolar macrophages exhibited defective resolution of neutrophilic inflammation, exuded protein, lung edema, and lung tissue injury after ventilation, whereas these delayed responses were reversed by administration of Tlr4+/+ BMDMs. Importantly, these proresolving effects by Tlr4+/+ BMDMs were abolished in mice receiving Tlr4-/- BMDMs. The number of macrophages containing apoptotic cells or bodies in bronchoalveolar lavage fluid was much less in mice receiving Tlr4-/- BMDMs than that in those receiving Tlr4+/+ BMDMs. Macrophage TLR4 deletion facilitated a disintegrin and metalloprotease 17 maturation and enhanced Mer cleavage in response to mechanical ventilation. Heat shock protein 70 dramatically increased Mer tyrosine kinase surface expression, phagocytosis of apoptotic neutrophils, and rescued the inflammatory phenotype in alveolar macrophage-depleted mice receiving Tlr4+/+ BMDMs, but not Tlr4-/- BMDMs. Our results suggest that macrophage TLR4 promotes resolution of VILI via modulation of Mer-mediated efferocytosis.


Macrophages, Alveolar/metabolism , Neutrophils/immunology , Phagocytosis/physiology , Toll-Like Receptor 4/metabolism , Ventilator-Induced Lung Injury/pathology , ADAM17 Protein/metabolism , Animals , Apoptosis/physiology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Cells, Cultured , Female , HSP70 Heat-Shock Proteins/metabolism , Lung/pathology , Macrophages, Alveolar/transplantation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Respiration, Artificial/adverse effects , Signal Transduction , c-Mer Tyrosine Kinase/metabolism
3.
Am J Physiol Lung Cell Mol Physiol ; 321(4): L686-L702, 2021 10 01.
Article En | MEDLINE | ID: mdl-34318714

Increased lung vascular permeability and neutrophilic inflammation are hallmarks of acute lung injury. Alveolar macrophages (AMϕ), the predominant sentinel cell type in the airspace, die in massive numbers while fending off pathogens. Recent studies indicate that the AMϕ pool is replenished by airspace-recruited monocytes, but the mechanisms instructing the conversion of recruited monocytes into reparative AMϕ remain elusive. Cyclic AMP (cAMP) is a vascular barrier protective and immunosuppressive second messenger in the lung. Here, we subjected mice expressing GFP under the control of the Lysozyme-M promoter (LysM-GFP mice) to the LPS model of rapidly resolving lung injury to address the impact of mechanisms determining cAMP levels in AMϕ and regulation of mobilization of the reparative AMϕ-pool. RNA-seq analysis of flow-sorted Mϕ identified phosphodiesterase 4b (PDE4b) as the top LPS-responsive cAMP-regulating gene. We observed that PDE4b expression markedly increased at the time of peak injury (4 h) and then decreased to below the basal level during the resolution phase (24 h). Activation of transcription factor NFATc2 was required for the transcription of PDE4b in Mϕ. Inhibition of PDE4 activity at the time of peak injury, using intratracheal rolipram, increased cAMP levels, augmented the reparative AMϕ pool, and resolved lung injury. This response was not seen following conditional depletion of monocytes, thus establishing airspace-recruited PDE4b-sensitive monocytes as the source of reparative AMϕ. Interestingly, adoptive transfer of rolipram-educated AMϕ into injured mice resolved lung edema. We propose suppression of PDE4b as an effective approach to promote reparative AMϕ generation from monocytes for lung repair.


Acute Lung Injury/pathology , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Macrophages, Alveolar/cytology , Monocytes/cytology , NFATC Transcription Factors/metabolism , Adoptive Transfer/methods , Animals , Capillary Permeability/physiology , Cell Differentiation/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Female , Inflammation , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/transplantation , Male , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Phosphodiesterase 4 Inhibitors/pharmacology , Rolipram/pharmacology , Transcriptional Activation/genetics
4.
Sci Immunol ; 5(52)2020 Oct 23.
Article En | MEDLINE | ID: mdl-33097591

Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease in which airway macrophages (AMs) play a key role. Itaconate has emerged as a mediator of macrophage function, but its role during fibrosis is unknown. Here, we reveal that itaconate is an endogenous antifibrotic factor in the lung. Itaconate levels are reduced in bronchoalveolar lavage, and itaconate-synthesizing cis-aconitate decarboxylase expression (ACOD1) is reduced in AMs from patients with IPF compared with controls. In the murine bleomycin model of pulmonary fibrosis, Acod1−/− mice develop persistent fibrosis, unlike wild-type (WT) littermates. Profibrotic gene expression is increased in Acod1−/− tissue-resident AMs compared with WT, and adoptive transfer of WT monocyte-recruited AMs rescued mice from disease phenotype. Culture of lung fibroblasts with itaconate decreased proliferation and wound healing capacity, and inhaled itaconate was protective in mice in vivo. Collectively, these data identify itaconate as critical for controlling the severity of lung fibrosis, and targeting this pathway may be a viable therapeutic strategy.


Carboxy-Lyases/metabolism , Idiopathic Pulmonary Fibrosis/immunology , Macrophages, Alveolar/immunology , Succinates/metabolism , Administration, Inhalation , Adoptive Transfer/methods , Adult , Aged , Animals , Bleomycin/administration & dosage , Bleomycin/toxicity , Bronchoalveolar Lavage Fluid/immunology , Bronchoscopy , Case-Control Studies , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts , Healthy Volunteers , Humans , Hydro-Lyases/genetics , Hydro-Lyases/metabolism , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/diagnosis , Idiopathic Pulmonary Fibrosis/therapy , Lung/cytology , Lung/immunology , Lung/pathology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/transplantation , Male , Mice , Mice, Knockout , Middle Aged , Primary Cell Culture , Severity of Illness Index , Succinates/administration & dosage , Succinates/immunology
5.
Sci Immunol ; 5(50)2020 08 07.
Article En | MEDLINE | ID: mdl-32769172

Alveolar macrophages (AMs) are the major lung-resident macrophages and have contradictory functions. AMs maintain tolerance and tissue homeostasis, but they also initiate strong inflammatory responses. However, such opposing roles within the AM population were not known to be simultaneously generated and coexist. Here, we uncovered heterogeneous AM subpopulations generated in response to two distinct pulmonary fungal infections, Cryptococcus neoformans and Aspergillus fumigatus Some AMs are bona fide sentinel cells that produce chemoattractant CXCL2, which also serves as a marker for AM heterogeneity, in the context of pulmonary fungal infections. However, other AMs do not produce CXCL2 and other pro-inflammatory molecules. Instead, they highly produce anti-inflammatory molecules, including interleukin-10 (IL-10) and complement component 1q (C1q). These two AM subpopulations have distinct metabolic profiles and phagocytic capacities. We report that polarization of pro-inflammatory and anti-inflammatory AM subpopulations is regulated at both epigenetic and transcriptional levels and that these AM subpopulations are generally highly plastic. Our studies have uncovered the role of C1q expression in programming and sustaining anti-inflammatory AMs. Our finding of the AM heterogeneity upon fungal infections suggests a possible pharmacological intervention target to treat fungal infections by tipping the balance of AM subpopulations.


Aspergillosis/immunology , Aspergillus fumigatus , Chemokine CXCL2/immunology , Cryptococcosis/immunology , Macrophages, Alveolar/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Chemokine CXCL2/genetics , Female , Lung/immunology , Macrophages, Alveolar/transplantation , Male , Mice, Transgenic
6.
Mol Ther ; 27(9): 1597-1611, 2019 09 04.
Article En | MEDLINE | ID: mdl-31326401

Hereditary pulmonary alveolar proteinosis (PAP) is a genetic lung disease characterized by surfactant accumulation and respiratory failure arising from disruption of GM-CSF signaling. While mutations in either CSF2RA or CSF2RB (encoding GM-CSF receptor α or ß chains, respectively) can cause PAP, α chain mutations are responsible in most patients. Pulmonary macrophage transplantation (PMT) is a promising new cell therapy in development; however, no studies have evaluated this approach for hereditary PAP (hPAP) caused by Csf2ra mutations. Here, we report on the preclinical safety, tolerability, and efficacy of lentiviral-vector (LV)-mediated Csf2ra expression in macrophages and PMT of gene-corrected macrophages (gene-PMT therapy) in Csf2ra gene-ablated (Csf2ra-/-) mice. Gene-PMT therapy resulted in a stable transgene integration and correction of GM-CSF signaling and functions in Csf2ra-/- macrophages in vitro and in vivo and resulted in engraftment and long-term persistence of gene-corrected macrophages in alveoli; restoration of pulmonary surfactant homeostasis; correction of PAP-specific cytologic, histologic, and biomarker abnormalities; and reduced inflammation associated with disease progression in untreated mice. No adverse consequences of gene-PMT therapy in Csf2ra-/- mice were observed. Results demonstrate that gene-PMT therapy of hPAP in Csf2ra-/- mice was highly efficacious, durable, safe, and well tolerated.


Cell- and Tissue-Based Therapy , Genetic Therapy , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/transplantation , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Alveolar Proteinosis/therapy , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Animals , Cell Proliferation , Cell- and Tissue-Based Therapy/methods , Disease Models, Animal , Gene Expression , Genetic Therapy/methods , Genetic Vectors/genetics , Immunophenotyping , Lentivirus/genetics , Mice , Mice, Knockout , Pulmonary Alveolar Proteinosis/diagnosis , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction , Transduction, Genetic
7.
Am J Respir Cell Mol Biol ; 61(6): 727-736, 2019 12.
Article En | MEDLINE | ID: mdl-31162951

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive lung disorder and lacks effective treatments because of unclear mechanisms. Aberrant function of alveolar macrophages is directly linked to pulmonary fibrosis. Here, we show TIM-3 (T-cell immunoglobulin domain and mucin domain-3), a key regulator of macrophage function, aggravates pulmonary fibrosis. TIM-3 mRNA of patients with IPF was analyzed based on the Gene Expression Omnibus and Array Express databases. Lung pathology and profibrotic molecules were assessed in a bleomycin (BLM)-induced pulmonary fibrosis model using wild-type (WT) and TIM-3 transgenic (TIM-3-TG) mice. Macrophage cells, RAW264.7, were then applied to investigate the effect of macrophage TIM-3 under BLM exposure in vitro. Macrophage depletion and adoptive-transfer experiments were finally performed to examine lung morphology and profibrotic molecules. TIM-3 expression was increased both in patients with IPF and in our BLM-induced mouse model. TIM-3-TG mice developed more serious pathological changes in lung tissue and higher expressions of TGF-ß1 (transforming growth factor-ß1) and IL-10 than WT mice. After BLM treatment, TGF-ß1 and IL-10 expression was significantly decreased in RAW264.7 cells after TIM-3 knock-out, whereas it was increased in TIM-3-TG peritoneal macrophages. The scores of pulmonary fibrosis in WT and TIM-3-TG mice were significantly reduced, and there was no difference between them after macrophage depletion. Furthermore, WT mice receiving adoptive macrophages from TIM-3-TG mice also had more serious lung fibrosis and increased expression of TGF-ß1 and IL-10 than those receiving macrophages from WT mice. Our findings revealed that overexpressed TIM-3 in alveolar macrophages aggravated pulmonary fibrosis.


Hepatitis A Virus Cellular Receptor 2/blood , Hepatitis A Virus Cellular Receptor 2/physiology , Idiopathic Pulmonary Fibrosis/pathology , Macrophages, Alveolar/metabolism , Adoptive Transfer , Animals , Bleomycin/toxicity , CRISPR-Cas Systems , Disease Models, Animal , Hepatitis A Virus Cellular Receptor 2/deficiency , Hepatitis A Virus Cellular Receptor 2/genetics , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics , Interleukin-10/biosynthesis , Lung/pathology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/transplantation , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RAW 264.7 Cells , RNA, Messenger/biosynthesis , Transforming Growth Factor beta1/biosynthesis
8.
Curr Opin Pulm Med ; 25(3): 294-299, 2019 05.
Article En | MEDLINE | ID: mdl-30865035

PURPOSE OF REVIEW: Pulmonary alveolar proteinosis (PAP) is characterized by the massive accumulation of lipoproteinaceous material within alveoli, which results in progressive respiratory failure. The abnormalities in surfactant clearance are caused by defective pulmonary macrophages, whose terminal differentiation is GM-CSF-dependent. In hereditary PAP, the rupture of GM-CSF signaling is because of mutations in the GM-CSF receptor genes. This review focus on the innovative technologies of gene-correction proposed for the development of new therapeutic strategies, for hereditary PAP patients. RECENT FINDINGS: Hematopoietic stem cell gene therapy has been successfully experimented in murine models to restore the expression of the GM-CSF receptor, however, a therapeutic approach based on bone marrow transplantation requires a preconditioning, which could be hazardous in PAP patients, who are highly susceptible to pulmonary infections. Gene-corrected pulmonary macrophages, administered directly to the lung, could represent an improved approach. Finally, patient-derived induced pluripotent stem cells seem to be promising to overcome the limited availability of primary patient cells and to generate gene-corrected macrophages, able to recover pulmonary surfactant clearance. SUMMARY: WLL is the gold standard therapy for PAP. However, its use in hereditary PAP is limited by the difficulty of performing this technique in paediatric patients and by its purely symptomatic efficacy. The recent advances in genome engineering could provide efficacious strategies for clinical application.


Genetic Therapy , Macrophages, Alveolar/transplantation , Pluripotent Stem Cells/transplantation , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Alveolar Proteinosis/therapy , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Animals , Hematopoietic Stem Cell Transplantation , Humans , Pulmonary Surfactants
9.
JCI Insight ; 4(4)2019 02 21.
Article En | MEDLINE | ID: mdl-30668546

Allergic eosinophilic asthma is a chronic condition causing airway remodeling resulting in lung dysfunction. We observed that expression of sirtuin 2 (Sirt2), a histone deacetylase, regulates the recruitment of eosinophils after sensitization and challenge with a triple antigen: dust mite, ragweed, and Aspergillus fumigatus (DRA). Our data demonstrate that IL-4 regulates the expression of Sirt2 isoform 3/5. Pharmacological inhibition of Sirt2 by AGK2 resulted in diminished cellular recruitment, decreased CCL17/TARC, and reduced goblet cell hyperplasia. YM1 and Fizz1 expression was reduced in AGK2-treated, IL-4-stimulated lung macrophages in vitro as well as in lung macrophages from AGK2-DRA-challenged mice. Conversely, overexpression of Sirt2 resulted in increased cellular recruitment, CCL17 production, and goblet cell hyperplasia following DRA challenge. Sirt2 isoform 3/5 was upregulated in primary human alveolar macrophages following IL-4 and AGK2 treatment, which resulted in reduced CCL17 and markers of alternative activation. These gain-of-function and loss-of-function studies indicate that Sirt2 could be developed as a treatment for eosinophilic asthma.


Asthma/immunology , Eosinophils/immunology , Interleukin-4/immunology , Sirtuin 2/immunology , Adoptive Transfer , Allergens/administration & dosage , Allergens/immunology , Animals , Asthma/diagnosis , Asthma/pathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cells, Cultured , Chemokine CCL17/immunology , Chemokine CCL17/metabolism , Disease Models, Animal , Female , Furans/pharmacology , Goblet Cells/immunology , Goblet Cells/pathology , Humans , Interleukin-4/metabolism , Lung/cytology , Lung/immunology , Lung/pathology , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/transplantation , Male , Mice , Mice, Knockout , Primary Cell Culture , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , Quinolines/pharmacology , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sirtuin 2/antagonists & inhibitors , Sirtuin 2/genetics , Sirtuin 2/metabolism
10.
Shock ; 51(3): 389-400, 2019 03.
Article En | MEDLINE | ID: mdl-29608552

BACKGROUND: Acute lung injury (ALI) is a severe outcome of sepsis. Alveolar macrophages (AMs) play key roles in defense, resolution in ALI. The polarization of AMs is dependent on micro environmental stimuli and might influence the progression of ALI. Gainesville Tokushima scientists (GTS)-21, a selective α7 nicotinic acetylcholine receptor agonist of the cholinergic anti-inflammatory pathway (CAP), has recently been established to be promising in the treatment of ALI. However, the molecular mechanism underlying the GTS-21-mediated suppression of inflammatory responses has been explored only partially. In this study, we examined the relation between GTS-21 and AM polarization in ALI. METHODS: The adoptive transfer of M1 (classically activated) and M2 (alternatively activated)-polarized AMs was performed to AM-depleted ALI mice, along with the administration of GTS-21 in a murine model of lipopolysaccharide (LPS)-induced ALI and in isolated AMs that had been stimulated by LPS in vitro. RESULTS: The adoptive transfer of M1-polarized AMs aggravated the inflammatory response in the lung in contrast to the adoptive transfer of M2-polarized AMs. GTS-21 protected the lung from the effect of LPS, preventing injury and decreasing the number of AMs, AM-related pro-inflammatory cytokine levels, high mobility group box 1 expression levels in AMs. In addition, GTS-21 significantly diminished the number of M1-polarized AM and increased the number of M2-polarized AM, by flow cytometry, RT-PCR, enzyme-linked immunosorbent assay, and the Arg1 and iNOS activity assays. CONCLUSION: The GTS-21 substantially ameliorates LPS-induced ALI. This protection is predominantly associated with the inhibition of pulmonary AM M1 polarization and alteration in AM function.


Acute Lung Injury/therapy , Adoptive Transfer , Benzylidene Compounds/pharmacology , Macrophages, Alveolar/immunology , Pyridines/pharmacology , Sepsis/therapy , Acute Lung Injury/chemically induced , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Animals , Inflammation , Lipopolysaccharides/toxicity , Macrophages, Alveolar/pathology , Macrophages, Alveolar/transplantation , Mice , Nitric Oxide Synthase Type II/immunology , Sepsis/chemically induced , Sepsis/immunology , Sepsis/pathology
11.
Cell Rep ; 24(7): 1738-1746, 2018 08 14.
Article En | MEDLINE | ID: mdl-30110631

The rapid transit from hypoxia to normoxia in the lung that follows the first breath in newborn mice coincides with alveolar macrophage (AM) differentiation. However, whether sensing of oxygen affects AM maturation and function has not been previously explored. We have generated mice whose AMs show a deficient ability to sense oxygen after birth by deleting Vhl, a negative regulator of HIF transcription factors, in the CD11c compartment (CD11cΔVhl mice). VHL-deficient AMs show an immature-like phenotype and an impaired self-renewal capacity in vivo that persists upon culture ex vivo. VHL-deficient phenotype is intrinsic in AMs derived from monocyte precursors in mixed bone marrow chimeras. Moreover, unlike control Vhlfl/fl, AMs from CD11cΔVhl mice do not reverse pulmonary alveolar proteinosis when transplanted into Csf2rb-/- mice, demonstrating that VHL contributes to AM-mediated surfactant clearance. Thus, our results suggest that optimal AM terminal differentiation, self-renewal, and homeostatic function requires their intact oxygen-sensing capacity.


Cell Differentiation/genetics , Cell Proliferation/genetics , Hypoxia/genetics , Macrophages, Alveolar/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Animals , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD11 Antigens/genetics , CD11 Antigens/metabolism , CD11b Antigen/genetics , CD11b Antigen/metabolism , Cytokine Receptor Common beta Subunit/deficiency , Cytokine Receptor Common beta Subunit/genetics , Gene Deletion , Gene Expression Regulation , Humans , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung/metabolism , Lung/pathology , Macrophages, Alveolar/pathology , Macrophages, Alveolar/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen/pharmacology , Receptors, IgG/genetics , Receptors, IgG/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins , Signal Transduction , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
12.
Stem Cell Reports ; 11(3): 696-710, 2018 09 11.
Article En | MEDLINE | ID: mdl-30100408

Induced pluripotent stem cell (iPSC)-derived hematopoietic cells represent a highly attractive source for cell and gene therapy. Given the longevity, plasticity, and self-renewal potential of distinct macrophage subpopulations, iPSC-derived macrophages (iPSC-Mφ) appear of particular interest in this context. We here evaluated the airway residence, plasticity, and therapeutic efficacy of iPSC-Mφ in a murine model of hereditary pulmonary alveolar proteinosis (herPAP). We demonstrate that single pulmonary macrophage transplantation (PMT) of 2.5-4 × 106 iPSC-Mφ yields efficient airway residence with conversion of iPSC-Mφ to an alveolar macrophage (AMφ) phenotype characterized by a distinct surface marker and gene expression profile within 2 months. Moreover, PMT significantly improves alveolar protein deposition and other critical herPAP disease parameters. Thus, our data indicate iPSC-Mφ as a source of functional macrophages displaying substantial plasticity and therapeutic potential that upon pulmonary transplantation will integrate into the lung microenvironment, adopt an AMφ phenotype and gene expression pattern, and profoundly ameliorate pulmonary disease phenotypes.


Cytokine Receptor Common beta Subunit/genetics , Induced Pluripotent Stem Cells/cytology , Macrophages, Alveolar/cytology , Macrophages, Alveolar/transplantation , Pulmonary Alveolar Proteinosis/therapy , Animals , Cells, Cultured , Gene Deletion , Hematopoiesis , Mice , Mice, Knockout , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Alveolar Proteinosis/pathology
13.
J Immunol ; 201(2): 573-582, 2018 07 15.
Article En | MEDLINE | ID: mdl-29848754

Although influenza virus infection remains a concerning disease for public health, the roles of individual cytokines during the immune response to influenza infection are not fully understood. We have identified IL-36γ as a key mediator of immune protection during both high- and low-pathogenesis influenza infection. Il36g mRNA is upregulated in the lung following influenza infection, and mice lacking IL-36γ have greatly increased morbidity and mortality upon infection with either H1N1 or H3N2 influenza. The increased severity of influenza infection in IL-36γ-knockout (KO) mice is associated with increased viral titers, higher levels of proinflammatory cytokines early in infection, and more diffuse pathologic conditions late in the disease course. Interestingly, the increased severity of disease in IL-36γ-KO mice correlates with a rapid loss of alveolar macrophages following infection. We find that the alveolar macrophages from naive IL-36γ-KO mice have higher expression of M2-like surface markers compared with wild-type (WT) mice and show increased apoptosis within 24 h of infection. Finally, transfer of WT alveolar macrophages to IL-36γ-KO mice restores protection against lethal influenza challenge to levels observed in WT mice. Together, these data identify a critical role for IL-36γ in immunity against influenza virus and demonstrate the importance of IL-36γ signaling for alveolar macrophage survival during infection.


Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/physiology , Influenza, Human/immunology , Interleukin-1/metabolism , Lung/pathology , Macrophages, Alveolar/physiology , Orthomyxoviridae Infections/immunology , Adoptive Transfer , Animals , Cell Survival , Cells, Cultured , Humans , Interleukin-1/genetics , Macrophages, Alveolar/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Up-Regulation , Virus Replication
14.
Mucosal Immunol ; 9(3): 702-17, 2016 05.
Article En | MEDLINE | ID: mdl-26376364

CD163 is a macrophage scavenger receptor with anti-inflammatory and pro-inflammatory functions. Here, we report that alveolar macrophages (AMΦs) from asthmatic subjects had reduced cell-surface expression of CD163, which suggested that CD163 might modulate the pathogenesis of asthma. Consistent with this, house dust mite (HDM)-challenged Cd163(-/-) mice displayed increases in airway eosinophils and mucous cell metaplasia (MCM). The increased airway eosinophils and MCM in HDM-challenged Cd163(-/-) mice were mediated by augmented CCL24 production and could be reversed by administration of a neutralizing anti-CCL24 antibody. A proteomic analysis identified the calcium-dependent binding of CD163 to Dermatophagoides pteronyssinus peptidase 1 (Der p1). Der p1-challenged Cd163(-/-) mice had the same phenotype as HDM-challenged Cd163(-/-) mice with increases in airway eosinophils, MCM and CCL24 production, while Der p1 induced CCL24 secretion by bone marrow-derived macrophages (BMMΦs) from Cd163(-/-) mice, but not BMMΦs from wild-type (WT) mice. Finally, airway eosinophils and bronchoalveolar lavage fluid CCL24 levels were increased in Der p1-challenged WT mice that received adoptively transferred AMΦ's from Cd163(-/-) mice. Thus, we have identified CD163 as a macrophage receptor that binds Der p1. Furthermore, we have shown that HDM-challenged Cd163(-/-) mice have increased eosinophilic airway inflammation and MCM that are mediated by a CCL24-dependent mechanism.


Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Asthma/immunology , Chemokine CCL24/metabolism , Eosinophils/immunology , Macrophages, Alveolar/immunology , Receptors, Cell Surface/metabolism , Respiratory Mucosa/pathology , Animals , Antibodies, Neutralizing/administration & dosage , Antigens, CD/genetics , Antigens, Dermatophagoides/immunology , Antigens, Dermatophagoides/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Arthropod Proteins/immunology , Arthropod Proteins/metabolism , Cell Movement , Cells, Cultured , Chemokine CCL24/immunology , Cysteine Endopeptidases/immunology , Cysteine Endopeptidases/metabolism , Humans , Macrophages, Alveolar/transplantation , Metaplasia , Mice , Mice, Inbred C57BL , Mice, Knockout , Pyroglyphidae , Receptors, Cell Surface/genetics
17.
Lasers Med Sci ; 30(4): 1357-65, 2015 May.
Article En | MEDLINE | ID: mdl-25794592

Gold-based nanoparticles have been used in a number of therapeutic and diagnostic applications. The purpose of this study was to investigate the efficacy of gold-silica nanoshells (AuNS) in photothermal therapy (PTT) of rat gliomas. Rat alveolar macrophages (Ma) were used as nanoparticle delivery vectors. Uptake of AuNS (bare and PEGylated) was investigated in Ma. AuNS were incubated with Ma for 24 h. Phase contrast microscopy was used to visualize the distribution of loaded Ma in three-dimensional glioma spheroids. PTT efficacy was evaluated for both empty (Ma) and AuNS-loaded Ma (Ma(NS)) in both monolayers and spheroids consisting of C6 rat glioma cells and Ma. Monolayers/spheroids were irradiated for 5 min with light from an 810-nm diode laser at irradiances ranging from 7 to 28 W cm(-2). Monolayer survival was evaluated using a 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay while PTT efficacy in spheroids was determined from growth kinetics and live/dead fluorescence microscopy. PTT efficacy was investigated in vivo using a Sprague-Dawley rat glioma model. Five rats received direct intracranial injection of a mixture of 10(4) C6 glioma cells and, 2 days later, an equal number of Ma(NS). Three rats received laser treatment (810 nm; 10 min; 1 W) while the remaining two served as controls (no laser treatment). The uptake ratio of bare to PEGylated AuNS by Ma was 4:1. A significant photothermal effect was observed in vitro, albeit at relatively high radiant exposures (2.1-4.2 kJ cm(-2)). PTT proved effective in vivo in preventing or delaying tumor development in the PTT-treated animals.


Brain Neoplasms/therapy , Glioma/therapy , Nanoshells/administration & dosage , Animals , Cell Line, Tumor , Gold/chemistry , Hyperthermia, Induced , Lasers, Semiconductor/therapeutic use , Macrophages, Alveolar/transplantation , Male , Nanoshells/chemistry , Phototherapy , Rats , Rats, Sprague-Dawley , Silicon Dioxide/chemistry , Treatment Outcome
18.
J Synchrotron Radiat ; 22(1): 143-55, 2015 Jan.
Article En | MEDLINE | ID: mdl-25537601

Functionalized computed tomography (CT) in combination with labelled cells is virtually non-existent due to the limited sensitivity of X-ray-absorption-based imaging, but would be highly desirable to realise cell tracking studies in entire organisms. In this study we applied in-line free propagation X-ray phase-contrast CT (XPCT) in an allergic asthma mouse model to assess structural changes as well as the biodistribution of barium-labelled macrophages in lung tissue. Alveolar macrophages that were barium-sulfate-loaded and fluorescent-labelled were instilled intratracheally into asthmatic and control mice. Mice were sacrificed after 24 h, lungs were kept in situ, inflated with air and scanned utilizing XPCT at the SYRMEP beamline (Elettra Synchrotron Light Source, Italy). Single-distance phase retrieval was used to generate data sets with ten times greater contrast-to-noise ratio than absorption-based CT (in our setup), thus allowing to depict and quantify structural hallmarks of asthmatic lungs such as reduced air volume, obstruction of airways and increased soft-tissue content. Furthermore, we found a higher concentration as well as a specific accumulation of the barium-labelled macrophages in asthmatic lung tissue. It is believe that XPCT will be beneficial in preclinical asthma research for both the assessment of therapeutic response as well as the analysis of the role of the recruitment of macrophages to inflammatory sites.


Barium Sulfate , Contrast Media , Lung/cytology , Macrophages, Alveolar/diagnostic imaging , Synchrotrons , Tomography, X-Ray Computed/instrumentation , Algorithms , Allergens/toxicity , Animals , Asthma/chemically induced , Asthma/diagnostic imaging , Asthma/pathology , Barium Sulfate/pharmacokinetics , Cell Line, Transformed , Cell Movement , Contrast Media/pharmacokinetics , Disease Models, Animal , Female , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Lung/diagnostic imaging , Macrophages, Alveolar/physiology , Macrophages, Alveolar/transplantation , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Ovalbumin/immunology , Ovalbumin/toxicity , Tomography, X-Ray Computed/methods
19.
Nature ; 514(7523): 450-4, 2014 Oct 23.
Article En | MEDLINE | ID: mdl-25274301

Bone-marrow transplantation is an effective cell therapy but requires myeloablation, which increases infection risk and mortality. Recent lineage-tracing studies documenting that resident macrophage populations self-maintain independently of haematological progenitors prompted us to consider organ-targeted, cell-specific therapy. Here, using granulocyte-macrophage colony-stimulating factor (GM-CSF) receptor-ß-deficient (Csf2rb(-/-)) mice that develop a myeloid cell disorder identical to hereditary pulmonary alveolar proteinosis (hPAP) in children with CSF2RA or CSF2RB mutations, we show that pulmonary macrophage transplantation (PMT) of either wild-type or Csf2rb-gene-corrected macrophages without myeloablation was safe and well-tolerated and that one administration corrected the lung disease, secondary systemic manifestations and normalized disease-related biomarkers, and prevented disease-specific mortality. PMT-derived alveolar macrophages persisted for at least one year as did therapeutic effects. Our findings identify mechanisms regulating alveolar macrophage population size in health and disease, indicate that GM-CSF is required for phenotypic determination of alveolar macrophages, and support translation of PMT as the first specific therapy for children with hPAP.


Cell Transplantation , Cytokine Receptor Common beta Subunit/genetics , Genetic Therapy , Lung/cytology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/transplantation , Pulmonary Alveolar Proteinosis/therapy , Animals , Cell Separation , Cytokine Receptor Common beta Subunit/deficiency , Female , Lung/metabolism , Lung/pathology , Male , Mice , Oligonucleotide Array Sequence Analysis , Phenotype , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Alveolar Proteinosis/pathology , Time Factors
...