Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.395
Filter
1.
Front Immunol ; 15: 1425466, 2024.
Article in English | MEDLINE | ID: mdl-39100672

ABSTRACT

Introduction: Genetic mutations in critical nodes of pulmonary epithelial function are linked to the pathogenesis of pulmonary fibrosis (PF) and other interstitial lung diseases. The slow progression of these pathologies is often intermitted and accelerated by acute exacerbations, complex non-resolving cycles of inflammation and parenchymal damage, resulting in lung function decline and death. Excess monocyte mobilization during the initial phase of an acute exacerbation, and their long-term persistence in the lung, is linked to poor disease outcome. Methods: The present work leverages a clinical idiopathic PF dataset and a murine model of acute inflammatory exacerbations triggered by mutation in the alveolar type-2 cell-restricted Surfactant Protein-C [SP-C] gene to spatially and phenotypically define monocyte/macrophage changes in the fibrosing lung. Results: SP-C mutation triggered heterogeneous CD68+ macrophage activation, with highly active peri-injured cells relative to those sampled from fully remodeled and healthy regions. Ingenuity pathway analysis of sorted CD11b-SigF+CD11c+ alveolar macrophages defined asynchronous activation of extracellular matrix re-organization, cellular mobilization, and Apolipoprotein E (Apoe) signaling in the fibrosing lung. Cell-cell communication analysis of single cell sequencing datasets predicted pro-fibrogenic signaling (fibronectin/Fn1, osteopontin/Spp1, and Tgfb1) emanating from Trem2/TREM2 + interstitial macrophages. These cells also produced a distinct lipid signature from alveolar macrophages and monocytes, characterized by Apoe expression. Mono- and di-allelic genetic deletion of ApoE in SP-C mutant mice had limited impact on inflammation and mortality up to 42 day after injury. Discussion: Together, these results provide a detailed spatio-temporal picture of resident, interstitial, and monocyte-derived macrophages during SP-C induced inflammatory exacerbations and end-stage clinical PF, and propose ApoE as a biomarker to identify activated macrophages involved in tissue remodeling.


Subject(s)
Pulmonary Fibrosis , Animals , Mice , Humans , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/metabolism , Phenotype , Disease Models, Animal , Pulmonary Surfactant-Associated Protein C/genetics , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mutation , Macrophage Activation/genetics , Macrophage Activation/immunology , Apolipoproteins E/genetics , Male , Inflammation/immunology , Disease Progression , Macrophages/immunology , Macrophages/metabolism , Lung/pathology , Lung/immunology , Lung/metabolism , Mice, Inbred C57BL , Female , Monocytes/immunology , Monocytes/metabolism
2.
J Nanobiotechnology ; 22(1): 476, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39135064

ABSTRACT

BACKGROUND: Current needle-based vaccination for respiratory viruses is ineffective at producing sufficient, long-lasting local immunity in the elderly. Direct pulmonary delivery to the resident local pulmonary immune cells can create long-term mucosal responses. However, criteria for drug vehicle design rules that can overcome age-specific changes in immune cell functions have yet to be established. RESULTS: Here, in vivo charge-based nanoparticle (NP) uptake was compared in mice of two age groups (2- and 16-months) within the four notable pulmonary antigen presenting cell (APC) populations: alveolar macrophages (AM), interstitial macrophages (IM), CD103+ dendritic cells (DCs), and CD11b+ DCs. Both macrophage populations exhibited preferential uptake of anionic nanoparticles but showed inverse rates of phagocytosis between the AM and IM populations across age. DC populations demonstrated preferential uptake of cationic nanoparticles, which remarkably did not significantly change in the aged group. Further characterization of cell phenotypes post-NP internalization demonstrated unique surface marker expression and activation levels for each APC population, showcasing heightened DC inflammatory response to NP delivery in the aged group. CONCLUSION: The age of mice demonstrated significant preferences in the charge-based NP uptake in APCs that differed greatly between macrophages and DCs. Carefully balance of the targeting and activation of specific types of pulmonary APCs will be critical to produce efficient, age-based vaccines for the growing elderly population.


Subject(s)
Antigen-Presenting Cells , Dendritic Cells , Lung , Mice, Inbred C57BL , Nanoparticles , Phagocytosis , Animals , Nanoparticles/chemistry , Mice , Lung/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Antigen-Presenting Cells/immunology , Macrophages, Alveolar/metabolism , Polyethylene Glycols/chemistry , Aging , Female , Age Factors
3.
Part Fibre Toxicol ; 21(1): 32, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39135079

ABSTRACT

BACKGROUND: Alveolar macrophages (AMs) have been predicted to affect the pulmonary clearance of nanomaterials; however, their qualitative and quantitative roles are poorly understood. In this study, carbon black nanoparticles (CBNPs) were instilled into the lungs of Wistar rats at 30, 100, and 300 µg/rat. The concentrations of particles in organs, including the lung, lung-associated lymph nodes (LALN), liver, spleen, and kidney, were evaluated at days 0 (immediately after instillation), 1, 7, 28, 60, and 90 post-instillation. RESULTS: The results indicated a multimodal pulmonary clearance pattern for CBNPs: slow clearance until day 28, fast clearance from days 28 to 60, and slow clearance from days 60 to 90. To determine the mechanism of this unique clearance pattern, CBNPs were instilled into AM-depleted rats using clodronate liposomes (CLO). At 28 days after instillation, the CBNP levels in the lungs treated with CLO showed about 31% higher reduction than in normal rats. In addition, the concentration of CBNPs in LALN treated with CLO significantly increased on day 28, whereas in normal rats, no detectable levels were observed. CONCLUSIONS: This result highlights that the prolonged retention of poorly soluble NPs in the lung until day 28 is mediated by the phagocytosis of AMs, and the fast clearance between days 28-60 is due to the turnover time of AMs, estimated around 1-2 months after birth. Similarly, new generations of AMs mediate the slow phase between days 60 and 90. However, further studies are needed to understand the multimodal clearance mechanism and the modulation of pulmonary clearance of poorly soluble NPs.


Subject(s)
Lung , Macrophages, Alveolar , Nanoparticles , Rats, Wistar , Soot , Animals , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/drug effects , Soot/toxicity , Soot/chemistry , Lung/metabolism , Lung/drug effects , Male , Tissue Distribution , Rats , Particle Size , Clodronic Acid/administration & dosage , Metabolic Clearance Rate
4.
J Gene Med ; 26(7): e3718, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38979822

ABSTRACT

BACKGROUND: Icariin (ICA) inhibits inflammatory response in various diseases, but the mechanism underlying ICA treating airway inflammation in asthma needs further understood. We aimed to predict and validate the potential targets of ICA against asthma-associated airway inflammation using network pharmacology and experiments. METHODS: The ovalbumin-induced asthma-associated airway inflammation mice model was established. The effects of ICA were evaluated by behavioral, airway hyperresponsiveness, lung pathological changes, inflammatory cell and cytokines counts. Next, the corresponding targets of ICA were mined via the SEA, CTD, HERB, PharmMapper, Symmap database and the literature. Pubmed-Gene and GeneCards databases were used to screen asthma and airway inflammation-related targets. The overlapping targets were used to build an interaction network, analyze gene ontology and enrich pathways. Subsequently, flow cytometry, quantitative real-time PCR and western blotting were employed for validation. RESULTS: ICA alleviated the airway inflammation of asthma; 402 targets of ICA, 5136 targets of asthma and 4531 targets of airway inflammation were screened; 216 overlapping targets were matched and predicted ICA possesses the potential to modulate asthmatic airway inflammation by macrophage activation/polarization. Additionally, ICA decreased M1 but elevated M2. Potential targets that were disrupted by asthma inflammation were restored by ICA treatment. CONCLUSIONS: ICA alleviates airway inflammation in asthma by inhibiting the M1 polarization of alveolar macrophages, which is related to metabolic reprogramming. Jun, Jak2, Syk, Tnf, Aldh2, Aldh9a1, Nos1, Nos2 and Nos3 represent potential targets of therapeutic intervention. The present study enhances understanding of the anti-airway inflammation effects of ICA, especially in asthma.


Subject(s)
Asthma , Disease Models, Animal , Flavonoids , Macrophage Activation , Macrophages, Alveolar , Network Pharmacology , Animals , Asthma/drug therapy , Asthma/metabolism , Mice , Flavonoids/pharmacology , Flavonoids/therapeutic use , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/immunology , Macrophage Activation/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Cytokines/metabolism , Ovalbumin , Lung/pathology , Lung/drug effects , Lung/metabolism , Female
5.
Sci Adv ; 10(28): eadl4913, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38985878

ABSTRACT

The pathophysiology of silicosis is poorly understood, limiting development of therapies for those who have been exposed to the respirable particle. We explored mechanisms of silica-induced pulmonary fibrosis in human lung samples collected from patients with occupational exposure to silica and in a longitudinal mouse model of silicosis using multiple modalities including whole-lung single-cell RNA sequencing and histological, biochemical, and physiologic assessments. In addition to pulmonary inflammation and fibrosis, intratracheal silica challenge induced osteoclast-like differentiation of alveolar macrophages and recruited monocytes, driven by induction of the osteoclastogenic cytokine, receptor activator of nuclear factor κΒ ligand (RANKL) in pulmonary lymphocytes, and alveolar type II cells. Anti-RANKL monoclonal antibody treatment suppressed silica-induced osteoclast-like differentiation in the lung and attenuated pulmonary fibrosis. We conclude that silica induces differentiation of pulmonary osteoclast-like cells leading to progressive lung injury, likely due to sustained elaboration of bone-resorbing proteases and hydrochloric acid. Interrupting osteoclast-like differentiation may therefore constitute a promising avenue for moderating lung damage in silicosis.


Subject(s)
Cell Differentiation , Osteoclasts , Pulmonary Fibrosis , Silicon Dioxide , Silicosis , Silicon Dioxide/toxicity , Animals , Humans , Osteoclasts/metabolism , Osteoclasts/drug effects , Osteoclasts/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/metabolism , Mice , Silicosis/pathology , Silicosis/metabolism , Silicosis/etiology , Cell Differentiation/drug effects , RANK Ligand/metabolism , Disease Models, Animal , Male , Lung/pathology , Lung/metabolism , Lung/drug effects , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Macrophages, Alveolar/drug effects , Female
6.
Semin Immunopathol ; 46(3-4): 7, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39060761

ABSTRACT

The innate immune system exhibits features of memory, termed trained immunity, which promote faster and more robust responsiveness to heterologous challenges. Innate immune memory is sustained through epigenetic modifications, affecting gene accessibility, and promoting a tailored gene transcription for an enhanced immune response. Alterations in the epigenetic landscape are intertwined with metabolic rewiring. Here, we review the metabolic pathways that underscore the induction and maintenance of trained immunity, including glycolysis, oxidative phosphorylation, the tricarboxylic acid cycle, and amino acid and lipid metabolism. The intricate interplay of these pathways is pivotal for establishing innate immune memory in distinct cellular compartments. We explore in particular the case of resident lung alveolar macrophages. We propose that leveraging the memory of the innate immune system may present therapeutic potential. Specifically, targeting the metabolic programs of innate immune cells is an emerging strategy for clinical interventions, either to boost immune responses in immunosuppressed conditions or to mitigate maladaptive activation in hyperinflammatory diseases.


Subject(s)
Epigenesis, Genetic , Immunity, Innate , Immunologic Memory , Humans , Animals , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/immunology , Energy Metabolism , Metabolic Networks and Pathways , Lipid Metabolism , Trained Immunity
7.
Cell Rep ; 43(7): 114453, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38985677

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) infection, a major cause of hospital- and community-acquired pneumonia, still has a high mortality rate. Extracellular vesicles (EVs), as crucial mediators of intercellular communication, have a significant impact on infectious diseases. However, the role of EVs from alveolar macrophages (AMs) in MRSA pneumonia remains unclear. We report that AMs phagocytose MRSA and release more EVs in mice with MRSA pneumonia. EVs from AMs harboring phagocytosed MRSA exhibit significant proinflammatory effects and induce necroptosis by delivering tumor necrosis factor α (TNF-α) and miR-146a-5p. Mechanically, the upregulated miR-146a-5p in these EVs enhances the phosphorylation of RIPK1, RIPK3, and MLKL by targeting TNF receptor-associated factor 6 (TRAF6), thereby promoting TNF-α-induced necroptosis. The combination of a TNF-α antagonist and an miR-146a-5p antagomir effectively improves the outcomes of mice with MRSA pneumonia. Overall, we reveal the pronecrotic effect of EVs from MRSA-infected AMs and provide a promising target for the prevention and treatment of MRSA pneumonia.


Subject(s)
Extracellular Vesicles , Macrophages, Alveolar , Methicillin-Resistant Staphylococcus aureus , MicroRNAs , Necroptosis , Animals , Extracellular Vesicles/metabolism , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/microbiology , Mice , MicroRNAs/metabolism , MicroRNAs/genetics , Phagocytosis , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/pathology , Staphylococcal Infections/metabolism , Male , Humans
8.
Sci Rep ; 14(1): 14974, 2024 06 28.
Article in English | MEDLINE | ID: mdl-38951667

ABSTRACT

Bovine alveolar macrophages (AMs) defend the lungs against pathogens such as Mycobacterium bovis (M. bovis), the causative agent of bovine tuberculosis. However, little is known about the surface molecules expressed by bovine AMs and whether there is heterogeneity within the population. The purpose of this study was to characterise the bovine AM cell surface phenotype using flow cytometry. Bronchoalveolar lavage samples from four different calves were stained with a combination of antibodies against immune cell molecules prior to flow cytometric analysis. To assess the degree of expression, we considered the distribution and relative intensities of stained and unstained cells. We demonstrated that bovine AMs have high expression of CD172a, ADGRE1, CD206, and CD14, moderate expression of CD80, MHC II, CD1b, and CD40, low expression of CX3CR1 and CD86, and little or no expression of CD16 and CD26. Two distinct subsets of bovine AMs were identified based on CD163 expression. Subsequent analysis showed that the CD163+ subset had greater expression of other typical macrophage molecules compared to the CD163- subset, suggesting that these cells may perform different roles during infection. The characterisation of the uninfected bovine AM phenotype will provide a foundation for the examination of M. bovis-infected AMs.


Subject(s)
Antigens, CD , Antigens, Differentiation, Myelomonocytic , Macrophages, Alveolar , Receptors, Cell Surface , Animals , Cattle , Macrophages, Alveolar/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, CD/metabolism , Receptors, Cell Surface/metabolism , Phenotype , Mycobacterium bovis/immunology , Flow Cytometry , Tuberculosis, Bovine/metabolism , Tuberculosis, Bovine/immunology , Tuberculosis, Bovine/microbiology , Immunophenotyping , Bronchoalveolar Lavage Fluid
9.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-39000325

ABSTRACT

One of the most significant diseases in the swine business, porcine reproductive and respiratory syndrome virus (PRRSV) causes respiratory problems in piglets and reproductive failure in sows. The PRRSV nucleocapsid (N) protein is essential for the virus' assembly, replication, and immune evasion. Stages in the viral replication cycle can be impacted by interactions between the PRRSV nucleocapsid protein and the host protein components. Therefore, it is of great significance to explore the interaction between the PRRSV nucleocapsid protein and the host. Nevertheless, no information has been published on the network of interactions between the nucleocapsid protein and the host proteins in primary porcine alveolar macrophages (PAMs). In this study, 349 host proteins interacting with nucleocapsid protein were screened in the PRRSV-infected PAMs through a liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based proteomics approach. Bioinformatics analysis, which included gene ontology annotation, Kyoto Encyclopedia of Genes and Genomes database enrichment, and a protein-protein interaction (PPI) network, revealed that the host proteins interacting with PRRSV-N may be involved in protein binding, DNA transcription, metabolism, and innate immune responses. This study confirmed the interaction between the nucleocapsid protein and the natural immune-related proteins. Ultimately, our findings suggest that the nucleocapsid protein plays a pivotal role in facilitating immune evasion during a PRRSV infection. This study contributes to enhancing our understanding of the role played by the nucleocapsid protein in viral pathogenesis and virus-host interaction, thereby offering novel insights for the prevention and control of PRRS as well as the development of vaccines.


Subject(s)
Host-Pathogen Interactions , Macrophages, Alveolar , Nucleocapsid Proteins , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Protein Interaction Maps , Proteomics , Tandem Mass Spectrometry , Animals , Swine , Porcine respiratory and reproductive syndrome virus/metabolism , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/virology , Proteomics/methods , Nucleocapsid Proteins/metabolism , Porcine Reproductive and Respiratory Syndrome/metabolism , Porcine Reproductive and Respiratory Syndrome/virology , Tandem Mass Spectrometry/methods , Chromatography, Liquid , Computational Biology/methods , Gene Ontology
10.
Int Immunopharmacol ; 139: 112719, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39032470

ABSTRACT

Autophagy in alveolar macrophages (AMs) is an important mechanism for maintaining immune homeostasis and normal lung tissue function, and insufficient autophagy in AMs may mediate the development of sepsis-induced acute lung injury (SALI). Insufficient autophagy in AMs and the activation of the NLRP3 inflammasome were observed in a mouse model with SALI induced by cecal ligation and puncture (CLP), resulting in the release of a substantial quantity of proinflammatory factors and the formation of SALI. However, after andrographolide (AG) intervention, autophagy in AMs was significantly promoted, the activation of the NLRP3 inflammasome was inhibited, the release of proinflammatory factors and pyroptosis were suppressed, and SALI was then ameliorated. In the MH-S cell model stimulated with LPS, insufficient autophagy was discovered to promote the overactivation of the NLRP3 inflammasome. AG was found to significantly promote autophagy, inhibit the activation of the NLRP3 inflammasome, and attenuate the release of proinflammatory factors. The primary mechanism of AG promoting autophagy was to inhibit the activation of the PI3K/AKT/mTOR pathway by binding RAGE to the membrane. In addition, it inhibited the activation of the NLRP3 inflammasome to ameliorate SALI. Our findings suggest that AG promotes autophagy in AMs through the RAGE/PI3K/AKT/mTOR pathway to inhibit the activation of the NLRP3 inflammasome, remodel the functional homeostasis of AMs in SALI, and exert anti-inflammatory and lung-protective effects. It has also been the first to suggest that RAGE is likely a direct target through which AG regulates autophagy, providing theoretical support for a novel therapeutic strategy in sepsis.


Subject(s)
Acute Lung Injury , Autophagy , Diterpenes , Macrophages, Alveolar , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Receptor for Advanced Glycation End Products , Sepsis , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , TOR Serine-Threonine Kinases/metabolism , Autophagy/drug effects , Sepsis/drug therapy , Sepsis/complications , Sepsis/metabolism , Sepsis/immunology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Diterpenes/pharmacology , Diterpenes/therapeutic use , Mice , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Male , Receptor for Advanced Glycation End Products/metabolism , Inflammasomes/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal
11.
Nat Commun ; 15(1): 5730, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977695

ABSTRACT

The circular RNA (circRNA) family is a group of endogenous non-coding RNAs (ncRNAs) that have critical functions in multiple physiological and pathological processes, including inflammation, cancer, and cardiovascular diseases. However, their roles in regulating innate immune responses remain unclear. Here, we define Cell division cycle 42 (CDC42)-165aa, a protein encoded by circRNA circCDC42, which is overexpressed in Klebsiella pneumoniae (KP)-infected alveolar macrophages. High levels of CDC42-165aa induces the hyperactivation of Pyrin inflammasomes and aggravates alveolar macrophage pyroptosis, while the inhibition of CDC42-165aa reduces lung injury in mice after KP infection by inhibiting Pyrin inflammasome-mediated pyroptosis. Overall, these results demonstrate that CDC42-165aa stimulates Pyrin inflammasome by inhibiting CDC42 GTPase activation and provides a potential clinical target for pathogenic bacterial infection in clinical practice.


Subject(s)
Inflammasomes , Klebsiella Infections , Klebsiella pneumoniae , Mice, Inbred C57BL , Pyroptosis , cdc42 GTP-Binding Protein , Animals , Pyroptosis/genetics , Klebsiella Infections/immunology , Klebsiella Infections/microbiology , Klebsiella Infections/metabolism , Mice , Inflammasomes/metabolism , cdc42 GTP-Binding Protein/metabolism , cdc42 GTP-Binding Protein/genetics , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Male , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Humans , Immunity, Innate , Macrophages/metabolism , Macrophages/immunology , Macrophages/microbiology , CARD Signaling Adaptor Proteins
12.
Cell Mol Life Sci ; 81(1): 287, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38970705

ABSTRACT

Lung type 2 pneumocytes (T2Ps) and alveolar macrophages (AMs) play crucial roles in the synthesis, recycling and catabolism of surfactant material, a lipid/protein fluid essential for respiratory function. The liver X receptors (LXR), LXRα and LXRß, are transcription factors important for lipid metabolism and inflammation. While LXR activation exerts anti-inflammatory actions in lung injury caused by lipopolysaccharide (LPS) and other inflammatory stimuli, the full extent of the endogenous LXR transcriptional activity in pulmonary homeostasis is incompletely understood. Here, using mice lacking LXRα and LXRß as experimental models, we describe how the loss of LXRs causes pulmonary lipidosis, pulmonary congestion, fibrosis and chronic inflammation due to defective de novo synthesis and recycling of surfactant material by T2Ps and defective phagocytosis and degradation of excess surfactant by AMs. LXR-deficient T2Ps display aberrant lamellar bodies and decreased expression of genes encoding for surfactant proteins and enzymes involved in cholesterol, fatty acids, and phospholipid metabolism. Moreover, LXR-deficient lungs accumulate foamy AMs with aberrant expression of cholesterol and phospholipid metabolism genes. Using a house dust mite aeroallergen-induced mouse model of asthma, we show that LXR-deficient mice exhibit a more pronounced airway reactivity to a methacholine challenge and greater pulmonary infiltration, indicating an altered physiology of LXR-deficient lungs. Moreover, pretreatment with LXR agonists ameliorated the airway reactivity in WT mice sensitized to house dust mite extracts, confirming that LXR plays an important role in lung physiology and suggesting that agonist pharmacology could be used to treat inflammatory lung diseases.


Subject(s)
Homeostasis , Liver X Receptors , Macrophages, Alveolar , Pneumonia , Pulmonary Surfactants , Signal Transduction , Animals , Liver X Receptors/metabolism , Liver X Receptors/genetics , Pulmonary Surfactants/metabolism , Mice , Pneumonia/metabolism , Pneumonia/pathology , Macrophages, Alveolar/metabolism , Mice, Inbred C57BL , Mice, Knockout , Lung/metabolism , Lung/pathology , Alveolar Epithelial Cells/metabolism , Asthma/metabolism , Asthma/pathology , Asthma/genetics , Cholesterol/metabolism , Lipid Metabolism , Phagocytosis
13.
J Transl Med ; 22(1): 698, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075394

ABSTRACT

BACKGROUND: Severe COVID-19 infection has been associated with the development of pulmonary fibrosis, a condition that significantly affects patient prognosis. Understanding the underlying cellular communication mechanisms contributing to this fibrotic process is crucial. OBJECTIVE: In this study, we aimed to investigate the role of the TNFSF12-TNFRSF12A pathway in mediating communication between alveolar macrophages and fibroblasts, and its implications for the development of pulmonary fibrosis in severe COVID-19 patients. METHODS: We conducted single-cell RNA sequencing (scRNA-seq) analysis using lung tissue samples from severe COVID-19 patients and healthy controls. The data was processed, analyzed, and cell types were annotated. We focused on the communication between alveolar macrophages and fibroblasts and identified key signaling pathways. In vitro experiments were performed to validate our findings, including the impact of TNFRSF12A silencing on fibrosis reversal. RESULTS: Our analysis revealed that in severe COVID-19 patients, alveolar macrophages communicate with fibroblasts primarily through the TNFSF12-TNFRSF12A pathway. This communication pathway promotes fibroblast proliferation and expression of fibrotic factors. Importantly, silencing TNFRSF12A effectively reversed the pro-proliferative and pro-fibrotic effects of alveolar macrophages. CONCLUSION: The TNFSF12-TNFRSF12A pathway plays a central role in alveolar macrophage-fibroblast communication and contributes to pulmonary fibrosis in severe COVID-19 patients. Silencing TNFRSF12A represents a potential therapeutic strategy for mitigating fibrosis in severe COVID-19 lung disease.


Subject(s)
COVID-19 , Fibroblasts , Macrophages, Alveolar , Pulmonary Fibrosis , Signal Transduction , TWEAK Receptor , Humans , COVID-19/complications , COVID-19/pathology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/complications , TWEAK Receptor/metabolism , TWEAK Receptor/genetics , Cytokine TWEAK/metabolism , Cell Communication , Male , SARS-CoV-2 , Female , Middle Aged , Cell Proliferation , Lung/pathology , Severity of Illness Index
14.
Sci Adv ; 10(31): eadm8836, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39083602

ABSTRACT

In the pathogenesis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, epithelial populations in the distal lung expressing Angiotensin-converting enzyme 2 (ACE2) are infrequent, and therefore, the model of viral expansion and immune cell engagement remains incompletely understood. Using human lungs to investigate early host-viral pathogenesis, we found that SARS-CoV-2 had a rapid and specific tropism for myeloid populations. Human alveolar macrophages (AMs) reliably expressed ACE2 allowing both spike-ACE2-dependent viral entry and infection. In contrast to Influenza A virus, SARS-CoV-2 infection of AMs was productive, amplifying viral titers. While AMs generated new viruses, the interferon responses to SARS-CoV-2 were muted, hiding the viral dissemination from specific antiviral immune responses. The reliable and veiled viral depot in myeloid cells in the very early phases of SARS-CoV-2 infection of human lungs enables viral expansion in the distal lung and potentially licenses subsequent immune pathologies.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Lung , Macrophages, Alveolar , Myeloid Cells , SARS-CoV-2 , Humans , SARS-CoV-2/physiology , COVID-19/virology , COVID-19/immunology , Angiotensin-Converting Enzyme 2/metabolism , Lung/virology , Lung/immunology , Lung/pathology , Macrophages, Alveolar/virology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Myeloid Cells/virology , Myeloid Cells/metabolism , Myeloid Cells/immunology , Virus Internalization , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/immunology , Viral Tropism
15.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000201

ABSTRACT

Pulmonary alveolar proteinosis (PAP) is an ultra-rare disease caused by impaired pulmonary surfactant clearance due to the dysfunction of alveolar macrophages or their signaling pathways. PAP is categorized into autoimmune, congenital, and secondary PAP, with autoimmune PAP being the most prevalent. This article aims to present a comprehensive review of PAP classification, pathogenesis, clinical presentation, diagnostics, and treatment. The literature search was conducted using the PubMed database and a total of 67 articles were selected. The PAP diagnosis is usually based on clinical symptoms, radiological imaging, and bronchoalveolar lavage, with additional GM-CSF antibody tests. The gold standard for PAP treatment is whole-lung lavage. This review presents a summary of the most recent findings concerning pulmonary alveolar proteinosis, pointing out specific features that require further investigation.


Subject(s)
Pulmonary Alveolar Proteinosis , Pulmonary Alveolar Proteinosis/therapy , Pulmonary Alveolar Proteinosis/diagnosis , Pulmonary Alveolar Proteinosis/pathology , Humans , Bronchoalveolar Lavage , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Pulmonary Surfactants/metabolism , Pulmonary Surfactants/therapeutic use , Macrophages, Alveolar/metabolism
16.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000242

ABSTRACT

Acute lung injury (ALI) is a condition associated with acute respiratory failure, resulting in significant morbidity and mortality. It involves cellular changes such as disruption of the alveolar-capillary membrane, excessive neutrophil migration, and release of inflammatory mediators. Broncho-Vaxom® (BV), a lyophilized product containing cell membrane components derived from eight bacteria commonly found in the respiratory tract, is known for its potential to reduce viral and bacterial lung infections. However, the specific effect of BV on ALI has not been clearly defined. This study explored the preventive effects of BV and its underlying mechanisms in a lipopolysaccharide (LPS)-induced ALI mouse model. Oral BV (1 mg/kg) gavage was administered one hour before the intratracheal injection of LPS to evaluate its preventive effect on the ALI model. The pre-administration of BV significantly mitigates inflammatory parameters, including the production of inflammatory mediators, macrophage infiltration, and NF-κB activation in lung tissue, and the increase in inflammatory cells in bronchoalveolar lavage fluid (BALF). Moreover, BV (3 µg/mL) pretreatment reduced the expression of M1 macrophage markers, interleukins (IL-1ß, IL-6), tumor necrosis factor α, and cyclooxygenase-2, which are activated by LPS, in both mouse alveolar macrophage MH-S cells and human macrophage THP-1 cells. These findings showed that BV exhibits anti-inflammatory effects by suppressing inflammatory mediators through the NF-κB pathway, suggesting its potential to attenuate bronchial and pulmonary inflammation.


Subject(s)
Acute Lung Injury , Disease Models, Animal , Lipopolysaccharides , Animals , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/drug therapy , Mice , Humans , Inflammation/pathology , Inflammation/metabolism , Inflammation/drug therapy , Male , Cell Extracts/pharmacology , Cell Extracts/therapeutic use , NF-kappa B/metabolism , Bronchoalveolar Lavage Fluid , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/drug effects , Cytokines/metabolism , Inflammation Mediators/metabolism , Lung/pathology , Lung/metabolism , Lung/drug effects , Bacterial Lysates
17.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-39000284

ABSTRACT

African swine fever (ASF), caused by the African swine fever virus (ASFV), is one of the most important infectious diseases that cause high morbidity and mortality in pigs and substantial economic losses to the pork industry of affected countries due to the lack of effective vaccines. The need to develop alternative robust antiviral countermeasures, especially anti-ASFV agents, is of the utmost urgency. This study shows that fangchinoline (FAN), a bisbenzylisoquinoline alkaloid found in the roots of Stephania tetrandra of the family Menispermaceae, significantly inhibits ASFV replication in porcine alveolar macrophages (PAMs) at micromolar concentrations (IC50 = 1.66 µM). Mechanistically, the infection of ASFV triggers the AKT/mTOR/NF-κB signaling pathway. FAN significantly inhibits ASFV-induced activation of such pathways, thereby suppressing viral replication. Such a mechanism was confirmed using an AKT inhibitor MK2206 as it inhibited AKT phosphorylation and ASFV replication in PAMs. Altogether, the results suggest that the AKT/mTOR pathway could potentially serve as a treatment strategy for combating ASFV infection and that FAN could potentially emerge as an effective novel antiviral agent against ASFV infections and deserves further in vivo antiviral evaluations.


Subject(s)
African Swine Fever Virus , Antiviral Agents , Benzylisoquinolines , Macrophages, Alveolar , NF-kappa B , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Virus Replication , Animals , Macrophages, Alveolar/virology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Virus Replication/drug effects , African Swine Fever Virus/drug effects , African Swine Fever Virus/physiology , Swine , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , NF-kappa B/metabolism , Benzylisoquinolines/pharmacology , Antiviral Agents/pharmacology , African Swine Fever/virology , African Swine Fever/drug therapy , African Swine Fever/metabolism
18.
Front Immunol ; 15: 1371764, 2024.
Article in English | MEDLINE | ID: mdl-38983858

ABSTRACT

Introduction: Environmental exposures and experimental manipulations can alter the ontogenetic composition of tissue-resident macrophages. However, the impact of these alterations on subsequent immune responses, particularly in allergic airway diseases, remains poorly understood. This study aims to elucidate the significance of modified macrophage ontogeny resulting from environmental exposures on allergic airway responses to house dust mite (HDM) allergen. Methods: We utilized embryonic lineage labeling to delineate the ontogenetic profile of tissue-resident macrophages at baseline and following the resolution of repeated lipopolysaccharide (LPS)-induced lung injury. We investigated differences in house dust mite (HDM)-induced allergy to assess the influence of macrophage ontogeny on allergic airway responses. Additionally, we employed single-cell RNA sequencing (scRNAseq) and immunofluorescent staining to characterize the pulmonary macrophage composition, associated pathways, and tissue localization. Results: Our findings demonstrate that the ontogeny of homeostatic alveolar and interstitial macrophages is altered after the resolution from repeated LPS-induced lung injury, leading to the replacement of embryonic-derived by bone marrow-derived macrophages. This shift in macrophage ontogeny is associated with reduced HDM-induced allergic airway responses. Through scRNAseq and immunofluorescent staining, we identified a distinct subset of resident-derived interstitial macrophages expressing genes associated with allergic airway diseases, localized adjacent to terminal bronchi, and diminished by prior LPS exposure. Discussion: These results suggest a pivotal role for pulmonary macrophage ontogeny in modulating allergic airway responses. Moreover, our findings highlight the implications of prior environmental exposures in shaping future immune responses and influencing the development of allergies. By elucidating the mechanisms underlying these phenomena, this study provides valuable insights into potential therapeutic targets for allergic airway diseases and avenues for further research into immune modulation and allergic disease prevention.


Subject(s)
Macrophages, Alveolar , Transcriptome , Animals , Mice , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Pyroglyphidae/immunology , Respiratory Hypersensitivity/immunology , Lung/immunology , Disease Models, Animal , Mice, Inbred C57BL , Allergens/immunology , Lipopolysaccharides , Female , Hypersensitivity/immunology
19.
J Vis Exp ; (209)2024 Jul 12.
Article in English | MEDLINE | ID: mdl-39072623

ABSTRACT

Pulmonary macrophages exhibit a dose-dependent pattern in phagocytizing particles. Following engulfment, these macrophages are subsequently excreted with sputum, rendering macrophages and particles visible and quantifiable under light microscopy. Notably, elemental carbon within the mammalian body originates exclusively from external contaminants. Consequently, the carbon content in airway macrophages (CCAM) serves as a valid exposure biomarker, accurately estimating individual exposure to carbon-containing particulate matter (PM). This article delineates a protocol involving sputum collection, preservation, processing, slide preparation, and staining, as well as macrophage photo acquisition and analysis. After removing the macrophage nuclei, the proportion of cytoplasm area occupied by carbon particles (PCOC) was calculated to quantify carbon content in each macrophage. The results indicate an elevation in CCAM levels after exposure to carbon-containing PM. In summary, this non-invasive, precise, reliable, and standardized method enables the direct measurement of carbon particles within target cells and is utilized for large-scale quantification of individual CCAM through induced sputum.


Subject(s)
Carbon , Particulate Matter , Sputum , Particulate Matter/analysis , Particulate Matter/toxicity , Humans , Carbon/chemistry , Carbon/toxicity , Sputum/chemistry , Sputum/cytology , Sputum/metabolism , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/chemistry , Macrophages, Alveolar/cytology
20.
PLoS Pathog ; 20(7): e1012398, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39038050

ABSTRACT

Inflammasomes play pivotal roles in inflammation by processing and promoting the secretion of IL-1ß. Caspase-1 is involved in the maturation of IL-1ß and IL-18, while human caspase-4 specifically processes IL-18. Recent structural studies of caspase-4 bound to Pro-IL-18 reveal the molecular basis of Pro-IL-18 activation by caspase-4. However, the mechanism of caspase-1 processing of pro-IL-1ß and other IL-1ß-converting enzymes remains elusive. Here, we observed that swine Pro-IL-1ß (sPro-IL-1ß) exists as an oligomeric precursor unlike monomeric human Pro-IL-1ß (hPro-IL-1ß). Interestingly, Seneca Valley Virus (SVV) 3C protease cleaves sPro-IL-1ß to produce mature IL-1ß, while it cleaves hPro-IL-1ß but does not produce mature IL-1ß in a specific manner. When the inflammasome is blocked, SVV 3C continues to activate IL-1ß through direct cleavage in porcine alveolar macrophages (PAMs). Through molecular modeling and mutagenesis studies, we discovered that the pro-domain of sPro-IL-1ß serves as an 'exosite' with its hydrophobic residues docking into a positively charged 3C protease pocket, thereby directing the substrate to the active site. The cleavage of sPro-IL-1ß generates a monomeric and active form of IL-1ß, initiating the downstream signaling. Thus, these studies provide IL-1ß is an inflammatory sensor that directly detects viral protease through an independent pathway operating in parallel with host inflammasomes.


Subject(s)
3C Viral Proteases , Inflammasomes , Interleukin-1beta , Picornaviridae , Viral Proteins , Animals , Interleukin-1beta/metabolism , 3C Viral Proteases/metabolism , Swine , Humans , Viral Proteins/metabolism , Inflammasomes/metabolism , Inflammation/metabolism , Picornaviridae Infections/metabolism , Picornaviridae Infections/virology , Cysteine Endopeptidases/metabolism , Species Specificity , Macrophages, Alveolar/virology , Macrophages, Alveolar/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL