Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 360
Filter
1.
Cell ; 184(14): 3774-3793.e25, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34115982

ABSTRACT

Cytomegaloviruses (CMVs) have co-evolved with their mammalian hosts for millions of years, leading to remarkable host specificity and high infection prevalence. Macrophages, which already populate barrier tissues in the embryo, are the predominant immune cells at potential CMV entry sites. Here we show that, upon CMV infection, macrophages undergo a morphological, immunophenotypic, and metabolic transformation process with features of stemness, altered migration, enhanced invasiveness, and provision of the cell cycle machinery for viral proliferation. This complex process depends on Wnt signaling and the transcription factor ZEB1. In pulmonary infection, mouse CMV primarily targets and reprograms alveolar macrophages, which alters lung physiology and facilitates primary CMV and secondary bacterial infection by attenuating the inflammatory response. Thus, CMV profoundly perturbs macrophage identity beyond established limits of plasticity and rewires specific differentiation processes, allowing viral spread and impairing innate tissue immunity.


Subject(s)
Cytomegalovirus/physiology , Macrophages, Alveolar/virology , Animals , Antigen Presentation , Bystander Effect , Cell Cycle , Cell Line, Transformed , Cellular Reprogramming , Cytomegalovirus/pathogenicity , Cytomegalovirus/ultrastructure , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Green Fluorescent Proteins/metabolism , Lung/pathology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/ultrastructure , Mice, Inbred BALB C , Mice, Inbred C57BL , Phenotype , Stem Cells/pathology , Virus Replication/physiology , Wnt Signaling Pathway
2.
Int Immunopharmacol ; 96: 107621, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33872850

ABSTRACT

Acute lung injury (ALI) is a common clinical condition with a high mortality rate and no specific treatment is available. An excessive inflammatory response contributes to the development of ALI and accelerates its progression, and the NLRP3 inflammasome and NF-κB signaling pathways are key players in inflammation. Platycodin D has been reported to have anti-oxidant and anti-stress properties in various diseases. However, the effects of PLD in ALI has not been clearly demonstrated. The aim of this study was to investigate the therapeutic effects of PLD on ALI and its possible mechanism. Our study found that PLD pre-treatment attenuated lung histopathological injury in LPS-induced SD rats and reduced the levels of inflammatory cytokines and lung wet/dry ratio in bronchoalveolar lavage fluid (BALF). In addition, PLD modulate LPS-induced production of MDA, MPO, GSH, GSH-Px and CAT in lung tissue. In addition, PLD suppressed the activation of NLRP3 inflammatory microsomes and the NF-κB signaling pathway. Thus, our results suggest that PLD are protective against LPS-induced ALI by inhibiting NLRP3 and NF-κB signaling pathway.


Subject(s)
Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Saponins/pharmacology , Toll-Like Receptor 4/metabolism , Triterpenes/pharmacology , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Animals , Apoptosis Regulatory Proteins/metabolism , Caspase 1/metabolism , Cytokines/genetics , Cytokines/metabolism , Down-Regulation/drug effects , Edema/chemically induced , Edema/drug therapy , I-kappa B Proteins/metabolism , Inflammasomes , Lipopolysaccharides/toxicity , Macrophages, Alveolar/ultrastructure , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neoplasm Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Organ Size/drug effects , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Saponins/therapeutic use , Signal Transduction/drug effects , Triterpenes/therapeutic use
3.
Eur J Clin Invest ; 51(1): e13443, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33131070

ABSTRACT

BACKGROUND: To reveal detailed histopathological changes, virus distributions, immunologic properties and multi-omic features caused by SARS-CoV-2 in the explanted lungs from the world's first successful lung transplantation of a COVID-19 patient. MATERIALS AND METHODS: A total of 36 samples were collected from the lungs. Histopathological features and virus distribution were observed by optical microscope and transmission electron microscope (TEM). Immune cells were detected by flow cytometry and immunohistochemistry. Transcriptome and proteome approaches were used to investigate main biological processes involved in COVID-19-associated pulmonary fibrosis. RESULTS: The histopathological changes of the lung tissues were characterized by extensive pulmonary interstitial fibrosis and haemorrhage. Viral particles were observed in the cytoplasm of macrophages. CD3+ CD4- T cells, neutrophils, NK cells, γ/δ T cells and monocytes, but not B cells, were abundant in the lungs. Higher levels of proinflammatory cytokines iNOS, IL-1ß and IL-6 were in the area of mild fibrosis. Multi-omics analyses revealed a total of 126 out of 20,356 significant different transcription and 114 out of 8,493 protein expression in lung samples with mild and severe fibrosis, most of which were related to fibrosis and inflammation. CONCLUSIONS: Our results provide novel insight that the significant neutrophil/ CD3+ CD4- T cell/ macrophage activation leads to cytokine storm and severe fibrosis in the lungs of COVID-19 patient and may contribute to a better understanding of COVID-19 pathogenesis.


Subject(s)
COVID-19/pathology , Hemorrhage/pathology , Lung Transplantation , Lung/pathology , Lymph Nodes/pathology , Pulmonary Fibrosis/pathology , B-Lymphocytes/pathology , B-Lymphocytes/ultrastructure , B-Lymphocytes/virology , COVID-19/genetics , COVID-19/metabolism , COVID-19/surgery , Chromatography, Liquid , Flow Cytometry , Gene Expression Profiling , Humans , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Killer Cells, Natural/pathology , Killer Cells, Natural/ultrastructure , Killer Cells, Natural/virology , Lung/metabolism , Lung/ultrastructure , Lung/virology , Lymph Nodes/metabolism , Lymph Nodes/ultrastructure , Lymph Nodes/virology , Macrophages, Alveolar/pathology , Macrophages, Alveolar/ultrastructure , Macrophages, Alveolar/virology , Male , Middle Aged , Monocytes/pathology , Monocytes/ultrastructure , Monocytes/virology , Neutrophils/pathology , Neutrophils/ultrastructure , Neutrophils/virology , Nitric Oxide Synthase Type II/metabolism , Proteomics , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/surgery , RNA-Seq , SARS-CoV-2 , Severity of Illness Index , T-Lymphocytes/pathology , T-Lymphocytes/ultrastructure , T-Lymphocytes/virology , Tandem Mass Spectrometry
4.
Int Immunopharmacol ; 86: 106728, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32593159

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) infection causes severe systemic inflammation. Based on transcriptome sequencing data, a new cold-inducible RNA-binding protein (CIRBP) was identified, and its upregulated expression was detected in PRRSV-infected porcine alveolar macrophages (PAMs). However, the immunoregulatoryeffect of CIRBP in PRRSV infection remains unclear. In this study, we found that CIRBP, as an RNA-binging protein, migrates to the cytoplasm from the nucleus and exists in cytoplasmic stress granules under PRRSV infection. In addition, as a new pro-inflammatory factor, the overexpression of CIRBP promotes the expression of inflammatory cytokines and oxidative stress as showing the production of iNOS and ROS in PRRSV-infected cells, which contributes to the inflammatory response via the NF-κB pathway. Our findings suggested that CIRBP is involved in the regulation of PRRSV-induced inflammatory response.


Subject(s)
Inflammation/metabolism , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine respiratory and reproductive syndrome virus/immunology , RNA-Binding Proteins/immunology , Animals , Cells, Cultured , Cytokines/metabolism , Inflammation/complications , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/ultrastructure , Macrophages, Alveolar/virology , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress , Porcine Reproductive and Respiratory Syndrome/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Swine , Up-Regulation
5.
Toxicol Mech Methods ; 30(7): 477-489, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32345130

ABSTRACT

Multi-walled carbon nanotubes (MWCNTs) have industrial applications in the nanotechnology field. The physico-chemical properties of MWCNTs vary greatly depending on MWCNT manufacture and application. It has been pointed out that their needle shape and high durability are important factors that determine the biopersistence of fibers and can lead to inhalation toxicity or cytotoxicity. In this study, we prepared six suspensions of MWCNTs differing in diameter and length, and performed in vitro cell-based assays for 24 h using NR8383 rat alveolar macrophages. Rigid, needle-shaped MWCNTs with a large diameter (>50 µm) penetrated the cytoplasm and decreased cell survival without generating intracellular reactive oxygen species (ROS), significantly up-regulated many genes involved in inflammatory responses, response to oxidative stress and apoptosis, and extracellular matrix degradation. Bent MWCNTs with a small diameter (<20 µm) were phagocytosed in vacuole-like cellular compartments and decreased cell survival along with intracellular ROS generation. Straight, thin MWCNTs with a small diameter (<20 µm) caused a slight intracellular ROS generation but no decrease in cell viability. Some straight, long, and thin MWCNTs were found in the mitochondria and near the nuclei; however, no mutagenesis was observed. The in vitro cell-based assays showed high cytotoxicity of MWCNTs with a large diameter (>50 µm), moderate and low cytotoxicity of MWCNTs with a small diameter (<20 µm). These results suggested that the diameter of MWCNTs considerably contributes to their cytotoxicity.


Subject(s)
Macrophages, Alveolar/drug effects , Nanotubes, Carbon/toxicity , Phagocytosis , Animals , Cell Line , Cell Survival/drug effects , Cytokines/genetics , Cytokines/metabolism , Inflammation Mediators/metabolism , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/ultrastructure , Oxidative Stress/drug effects , Particle Size , Rats , Reactive Oxygen Species/metabolism , Transcriptome
6.
Alcohol ; 83: 57-65, 2020 03.
Article in English | MEDLINE | ID: mdl-31499142

ABSTRACT

Several conditions are marked by increased susceptibility to, and enhanced severity of, bacterial infections. Alcohol use disorder, one of these conditions, is known to predispose to bacterial pneumonia by suppressing the lung's innate immune system, and more specifically by disrupting critical alveolar macrophage (AM) functions. Recently, we established that chronic ethanol consumption also perturbs surfactant lipid homeostasis in the lung and that elevated concentrations of free fatty acids contribute to blocking essential AM functions, such as agonist-induced cytokine expression. In this study, we extend these observations by showing that elevated free fatty acid levels impair metabolic responses to lipopolysaccharide (LPS) in AMs. In particular, we show that the glycolytic reprogramming characteristic of LPS-stimulated AMs is blunted by the saturated fatty acid palmitate, whereas oleate, an unsaturated fatty acid, or ethanol alone, had no effect on this adaptive metabolic response. Additionally, we found that elevated concentrations of palmitate induced mitochondrial oxidative stress and that glycolytic reprogramming and cytokine production to LPS could be partially restored in AMs by either pharmacologically blocking palmitate entry into mitochondria or administering a mitochondrial-specific antioxidant. Taken together, these findings suggest that alcohol and elevated levels of saturated fatty acids conspire to impair pulmonary innate immunity by altering metabolic responses in AMs. Additionally, our findings suggest that targeting the mechanisms involved in fatty acid metabolism can restore pulmonary immunity and possibly limit bacterial pneumonia in individuals with alcohol use disorder.


Subject(s)
Ethanol/toxicity , Glycolysis/drug effects , Lipid Metabolism/drug effects , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/physiology , Animals , Cell Line , Cytokines/metabolism , Fatty Acids/metabolism , Immunity/drug effects , Immunity/physiology , Macrophages, Alveolar/ultrastructure , Mitochondria/metabolism , Oxidative Stress/drug effects , Palmitates/antagonists & inhibitors , Palmitates/metabolism , Palmitates/pharmacology , Rats
8.
Lung ; 197(6): 687-698, 2019 12.
Article in English | MEDLINE | ID: mdl-31732808

ABSTRACT

PURPOSE: There is currently no true macrophage cell line and in vitro experiments requiring these cells currently require mitogenic stimulation of a macrophage precursor cell line (THP-1) or ex vivo maturation of circulating primary monocytes. In this study, we characterise a human macrophage cell line, derived from THP-1 cells, and compare its phenotype to the THP-1 cells. METHODS: THP-1 cells with and without mitogenic stimulation were compared to the newly derived macrophage-like cell line (Daisy) using microscopy, flow cytometry, phagocytosis assays, antigen binding assays and gene microarrays. RESULTS: We show that the cell line grows predominantly in an adherent monolayer. A panel of antibodies were chosen to investigate the cell surface phenotype of these cells using flow cytometry. Daisy cells expressed more CD11c, CD80, CD163, CD169 and CD206, but less CD14 and CD11b compared with mitogen-stimulated THP-1 cells. Unlike stimulated THP-1 cells which were barely able to bind immune complexes, Daisy cells showed large amounts of immune complex binding. Finally, although not statistically significant, the phagocytic ability of Daisy cells was greater than mitogen-stimulated THP-1 cells, suggesting that the cell line is more similar to mature macrophages. CONCLUSIONS: The observed phenotype suggests that Daisy cells are a good model of human macrophages with a phenotype similar to human alveolar macrophages.


Subject(s)
Antigen-Antibody Complex/metabolism , Macrophages, Alveolar/metabolism , Phagocytosis/physiology , RNA, Messenger/metabolism , THP-1 Cells/metabolism , Antigens, CD , Antigens, Differentiation, Myelomonocytic , B7-1 Antigen , CD11 Antigens , CD11b Antigen , Cell Line , Flow Cytometry , Humans , Immunophenotyping , Integrin alpha Chains , Lectins, C-Type , Lipopolysaccharide Receptors , Macrophages, Alveolar/physiology , Macrophages, Alveolar/ultrastructure , Mannose Receptor , Mannose-Binding Lectins , Microscopy , Microscopy, Electron, Transmission , Mitogens , Receptors, Cell Surface , Sialic Acid Binding Ig-like Lectin 1 , THP-1 Cells/physiology , Tissue Array Analysis
9.
Int J Mol Med ; 44(4): 1309-1324, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31432098

ABSTRACT

Sepsis is a highly heterogeneous syndrome that is caused by a dysregulated host response to infection. The disproportionate inflammatory response to invasive infection is a triggering event inducing sepsis. The activation of inflammasomes in sepsis can amplify inflammatory responses. It has been reported that damaged mitochondria contribute to NACHT, LRR and PYD domains­containing protein 3 (NLRP3) inflammasome­related sepsis. Our previous study revealed that hydrogen (H2) exerts anti­inflammatory effects in sepsis but the detailed mechanism remains to be elucidated. In the present study, septic mice induced by cecal ligation and puncture (CLP) and macrophages induced by lipopolysaccharide (LPS) were used as models of sepsis in vivo and in vitro, respectively. An inducer and inhibitor of autophagy and the NLRP3 inflammasome were administered to investigate the detailed mechanism of action of H2 treatment in sepsis. The results demonstrated that LPS and ATP led to NLRP3 inflammasome pathway activation, excessive cytokine release, mitochondrial dysfunction and the activation of autophagy. CLP induced organ injury and NLRP3 pathway activation. H2 treatment ameliorated vital organ damage, the inflammatory response, mitochondrial dysfunction and NLRP3 pathway activation, and promoted autophagy in macrophages induced by LPS and in CLP mice. However, the inhibitor of autophagy and the inducer of NLRP3 reversed the protective effect of H2 against organ damage, the inflammatory response and mitochondrial dysfunction in vivo and in vitro. Collectively, the results demonstrated that H2 alleviated mitochondrial dysfunction and cytokine release via autophagy­mediated NLRP3 inflammasome inactivation.


Subject(s)
Autophagy , Hydrogen/metabolism , Inflammasomes/metabolism , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sepsis/metabolism , Animals , Cytokines/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Macrophages, Alveolar/ultrastructure , Male , Mice , Reactive Oxygen Species/metabolism , Sepsis/diagnosis , Sepsis/etiology , Sepsis/mortality
10.
Biomaterials ; 203: 31-42, 2019 05.
Article in English | MEDLINE | ID: mdl-30851491

ABSTRACT

Nanocellulose is a promising bio-nanomaterial with attractive properties suitable for multiple industrial applications. The increased use of nanocellulose may lead to occupational exposure and negative health outcomes. However, knowledge on its health effects is limited, and while nanocellulose exposure may induce acute inflammatory responses in the lung, the underlying mechanisms are unknown. Alveolar macrophages are key cells in alveolar particle clearance. Their activation and function may be affected by various particles. Here, we investigated the uptake of pristine cellulose nanocrystals (CNC), and their effects on alveolar macrophage polarization and biological function. CNC uptake enhanced the secretion of several cytokines but did not on its own induce a complete macrophage polarization. In presence of macrophage activators, such as LPS/IFNG and IL4/IL13, CNC exposure enhanced the expression of M1 phenotype markers and the secretion of pro-inflammatory cytokines and chemokines, while decreasing M2 markers. CNC exposure also affected the function of activated alveolar macrophages resulting in a prominent cytokine burst and altered phagocytic activity. In conclusion, CNC exposure may result in dysregulation of macrophage activation and function that are critical in inflammatory responses in the lung.


Subject(s)
Cellulose/chemistry , Cellulose/pharmacology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Nanoparticles/chemistry , Phagocytes/drug effects , Animals , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Macrophages, Alveolar/ultrastructure , Mice , Microscopy, Electron, Scanning , Phagocytes/metabolism , Phagocytes/ultrastructure , Phagocytosis/drug effects , Phenotype
11.
Arch Toxicol ; 93(4): 909-920, 2019 04.
Article in English | MEDLINE | ID: mdl-30759267

ABSTRACT

Potassium octatitanate (K2O·8TiO2, POT) fibers are used as an alternative to asbestos. Their shape and biopersistence suggest that they are possibly carcinogenic. However, inhalation studies have shown that respired POT fibers have little carcinogenic potential. We conducted a short-term study in which we administered POT fibers, and anatase and rutile titanium dioxide nanoparticles (a-nTiO2, r-nTiO2) to rats using intra-tracheal intra-pulmonary spraying (TIPS). We found that similarly to other materials, POT fibers were more toxic than non-fibrous nanoparticles of the same chemical composition, indicating that the titanium dioxide composition of POT fibers does not appear to account for their lack of carcinogenicity. The present report describes the results of the 3-week and 52-week interim killing of our current 2-year study of POT fibers, with MWCNT-7 as a positive control and r-nTiO2 as a non-fibrous titanium dioxide control. Male F344 rats were administered 0.5 ml vehicle, 62.5 µg/ml and 125 µg/ml r-nTiO2 and POT fibers, and 125 µg/ml MWCNT-7 by TIPS every other day for 2 weeks (eight doses: total doses of 0.25 and 0.50 mg/rat). At 1 year, POT and MWCNT-7 fibers induced significant increases in alveolar macrophage number, granulation tissue in the lung, bronchiolo-alveolar cell hyperplasia and thickening of the alveolar wall, and pulmonary 8-OHdG levels. The 0.5 mg POT- and the MWCNT-7-treated groups also had increased visceral and parietal pleura thickness, increased mesothelial cell PCNA labeling indices, and a few areas of visceral mesothelial cell hyperplasia. In contrast, in the r-nTiO2-treated groups, none of the measured parameters were different from the controls.


Subject(s)
Lung/drug effects , Nanoparticles/toxicity , Nanotubes, Carbon/toxicity , Pleura/drug effects , Titanium/toxicity , Animals , Inhalation Exposure , Lung/metabolism , Lung/pathology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/ultrastructure , Male , Organ Size/drug effects , Pleura/metabolism , Pleura/pathology , Rats, Inbred F344 , Tissue Distribution , Titanium/pharmacokinetics
12.
J Cell Physiol ; 234(5): 5953-5963, 2019 05.
Article in English | MEDLINE | ID: mdl-30467847

ABSTRACT

OBJECTIVE: Exposure to coal dust causes the development of coal worker's pneumoconiosis (CWP), which is associated with accumulating macrophages in the lower respiratory tract. This study was performed to investigate the effect of tumor necrosis factor-α (TNF-α)-tumor necrosis factor receptor (TNFR) signal pathway on autophagy and apoptosis of alveolar macrophages (AMs) in CWP. METHODS: AMs from controls exposed to coal dust and CWP patients were collected, in which expressions of TNF-α and TNFR1 were determined. Autophagy was observed by transmission electron microscopy, and apoptosis by light microscope and using terminal deoxynucleotidyl transferase dUTP nick-end labeling staining. AMs in CWP patients were treated with TNF-α or anti-TNF-α antibody. Besides, expressions of autophagy marker proteins, apoptosis-related factors, FAS, caspase-8, and receptor-interacting serine-threonine-protein kinase 3 (RIPK3) were determined by western Blot. Activities of caspase-3 and caspase-8 were determined by a fluorescence kit. Flow cytometry was applied to measure the expression of TNFR1 on the surface of the AM. RESULTS: TNF-α expression and TNFR1 expression on the surface of AM, as well as autophagy and apoptotic index were significantly increased in AMs of CWP patients. In response to the treatment of TNF-α, TNF-α expression and TNFR1 expression on the surface of AM as well as LC3I expression were increased, autophagy was decreased, and LC3, LC3II, Beclin1 and B-cell lymphoma 2 expressions decreased, whereas FAS expression and activity and expression of caspase-3 and caspase-8 increased, and apoptotic index increased. Moreover, the situations were reversed with the treatment of anti-TNF-α antibody. CONCLUSION: TNF-α-TNFR signal pathway was involved in the occurrence and development of CWP by activating FAS-caspase-8 and thus inhibiting autophagy while promoting apoptosis of AM.


Subject(s)
Anthracosis/metabolism , Apoptosis , Autophagy , Macrophages, Alveolar/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adult , Anthracosis/genetics , Anthracosis/immunology , Anthracosis/pathology , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Proteins/metabolism , Case-Control Studies , Cells, Cultured , Humans , Macrophages, Alveolar/immunology , Macrophages, Alveolar/ultrastructure , Male , Middle Aged , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/immunology , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
13.
Small ; 15(4): e1803776, 2019 01.
Article in English | MEDLINE | ID: mdl-30536849

ABSTRACT

Recently, second harmonic generation (SHG) nanomaterials have been generated that are efficiently employed in the classical (NIR) and extended (NIR-II) near infrared windows using a multiphoton microscope. The aim was to test bismuth ferrite harmonic nanoparticles (BFO-HNPs) for their ability to monitor pulmonary macrophages in mice. BFO-loaded MH-S macrophages are given intratracheally to healthy mice or BFO-HNPs are intranasally instilled in mice with allergic airway inflammation and lung sections of up to 100 µM are prepared. Using a two-photon-laser scanning microscope, it is shown that bright BFO-HNPs signals are detected from superficially localized cells as well as from deep within the lung tissue. BFO-HNPs are identified with an excellent signal-to-noise ratio and virtually no background signal. The SHG from the nanocrystals can be distinguished from the endogenous collagen-derived SHG around the blood vessels and bronchial structures. BFO-HNPs are primarily taken up by M2 alveolar macrophages in vivo. This SHG imaging approach provides novel information about the interaction of macrophages with cells and the extracellular matrix in lung disease as it is capable of visualizing and tracking NP-loaded cells at high resolution in thick tissues with minimal background fluorescence.


Subject(s)
Bismuth/chemistry , Ferric Compounds/chemistry , Macrophages, Alveolar/cytology , Nanoparticles/chemistry , Animals , Bronchoalveolar Lavage , Female , Macrophages, Alveolar/ultrastructure , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Microscopy, Electron , Nanoparticles/ultrastructure
14.
Part Fibre Toxicol ; 15(1): 45, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30424790

ABSTRACT

BACKGROUND: Given the tremendous potential for graphene quantum dots (QDs) in biomedical applications, a thorough understanding of the interaction of these materials with macrophages is essential because macrophages are one of the most important barriers against exogenous particles. Although the cytotoxicity and cellular uptake of graphene QDs were reported in previous studies, the interaction between nuclei and the internalized graphene QDs is not well understood. We thus systematically studied the nuclear uptake and related nuclear response associated with aminated graphene QDs (AG-QDs) exposure. RESULTS: AG-QDs showed modest 24-h inhibition to rat alveolar macrophages (NR8383), with a minimum inhibitory concentration (MIC) of 200 µg/mL. Early apoptosis was significantly increased by AG-QDs (100 and 200 µg/mL) exposure and played a major role in cell death. The internalization of AG-QDs was mainly via energy-dependent endocytosis, phagocytosis and caveolae-mediated endocytosis. After a 48-h clearance period, more than half of the internalized AG-QDs remained in the cellular cytoplasm and nucleus. Moreover, AG-QDs were effectively accumulated in nucleus and were likely regulated by two nuclear pore complexes genes (Kapß2 and Nup98). AG-QDs were shown to alter the morphology, area, viability and nuclear components of exposed cells. Significant cleavage and cross-linking of DNA chains after AG-QDs exposure were confirmed by atomic force microscopy investigation. Molecular docking simulations showed that H-bonding and π-π stacking were the dominant forces mediating the interactions between AG-QDs and DNA, and were the important mechanisms resulting in DNA chain cleavage. In addition, the generation of reactive oxygen species (ROS) (e.g., •OH), and the up-regulation of caspase genes also contributed to DNA cleavage. CONCLUSIONS: AG-QDs were internalized by macrophages and accumulated in nuclei, which further resulted in nuclear damage and DNA cleavage. It is demonstrated that oxidative damage, direct contact via H-bonding and π-π stacking, and the up-regulation of caspase genes are the primary mechanisms for the observed DNA cleavage by AG-QDs.


Subject(s)
Cell Nucleus/drug effects , DNA Cleavage/drug effects , Exocytosis/drug effects , Graphite/toxicity , Macrophages, Alveolar/drug effects , Quantum Dots/toxicity , Animals , Apoptosis/drug effects , Cell Culture Techniques , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cell Survival/drug effects , DNA/metabolism , Graphite/pharmacokinetics , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/ultrastructure , Molecular Docking Simulation , Particle Size , Rats
15.
Int J Nanomedicine ; 13: 7061-7077, 2018.
Article in English | MEDLINE | ID: mdl-30464459

ABSTRACT

BACKGROUND: Titanium dioxide nanoparticles have numerous applications, resulting in human exposure. Nonetheless, available toxicological and safety data are insufficient regarding aspherical particles, such as rod-shaped nanoparticles. METHODS: In a combined in vitro-in vivo approach, cultured A549 lung alveolar adenocarcinoma cells were treated with approximately 15×65 nm TiO2 nanorod-containing medium, while young adult rats received the same substance by intratracheal instillation for 28 days in 5 and 18 mg/kg body-weight doses. Nanoparticle accumulation in the lungs and consequent oxidative stress, cell damage, and inflammation were assessed by biochemical and histopathological methods. RESULTS: Titanium was detected in tissue samples by single-particle inductively coupled plasma mass spectrometry. Nanoparticles were visualized inside cultured A549 cells, within pulmonary macrophages, and in hilar lymph nodes of the rats. A549 cells showed dose-dependent oxidative stress and lethality, and the observed nanoparticle-laden endosomes suggested deranged lysosomal function and possible autophagy. Strongly elevated Ti levels were measured in the lungs of nanorod-treated rats and moderately elevated levels in the blood of the animals. Numerous cytokines, indicating acute and also chronic inflammation, were identified in the lung samples of TiO2-exposed rodents. CONCLUSION: Several signs of cell and tissue damage were detected in both the cultured alveolar cells and in treated rats' lungs. Rod-shaped nanoparticulate TiO2 may consequently be more harmful than has generally been supposed. The occupational health risk suggested by the results calls for improved safety measures.


Subject(s)
Alveolar Epithelial Cells/drug effects , Nanotubes/chemistry , Titanium/pharmacology , A549 Cells , Animals , Body Weight , Cell Survival/drug effects , Cytokines/metabolism , Endocytosis/drug effects , Humans , Kidney/drug effects , Kidney/metabolism , Lymph Nodes/drug effects , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/ultrastructure , Male , Nanotubes/ultrastructure , Organ Size , Oxidative Stress/drug effects , Particle Size , Rats, Wistar , Titanium/blood
16.
Nat Commun ; 9(1): 3333, 2018 08 20.
Article in English | MEDLINE | ID: mdl-30127354

ABSTRACT

Mucormycosis is a life-threatening respiratory fungal infection predominantly caused by Rhizopus species. Mucormycosis has incompletely understood pathogenesis, particularly how abnormalities in iron metabolism compromise immune responses. Here we show how, as opposed to other filamentous fungi, Rhizopus spp. establish intracellular persistence inside alveolar macrophages (AMs). Mechanistically, lack of intracellular swelling of Rhizopus conidia results in surface retention of melanin, which induces phagosome maturation arrest through inhibition of LC3-associated phagocytosis. Intracellular inhibition of Rhizopus is an important effector mechanism, as infection of immunocompetent mice with swollen conidia, which evade phagocytosis, results in acute lethality. Concordantly, AM depletion markedly increases susceptibility to mucormycosis. Host and pathogen transcriptomics, iron supplementation studies, and genetic manipulation of iron assimilation of fungal pathways demonstrate that iron restriction inside macrophages regulates immunity against Rhizopus. Our findings shed light on the pathogenetic mechanisms of mucormycosis and reveal the role of macrophage-mediated nutritional immunity against filamentous fungi.


Subject(s)
Host-Pathogen Interactions , Iron/metabolism , Lung/microbiology , Macrophages, Alveolar/metabolism , Rhizopus/physiology , Animals , Cell Wall/metabolism , Gene Expression Regulation , Macrophages, Alveolar/ultrastructure , Melanins/metabolism , Mice, Inbred C57BL , Microbial Viability , Models, Biological , Mucormycosis/genetics , Mucormycosis/microbiology , Mucormycosis/pathology , Phagosomes/metabolism , Phagosomes/ultrastructure , Rhizopus/growth & development , Spores, Fungal/physiology
17.
Toxicol In Vitro ; 52: 351-364, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30031032

ABSTRACT

Considering that cigarette smoke condensate (CSC) is primarily absorbed through the alveoli in the lungs, herein, we used a mouse alveolar macrophage cell line (MH-S cells). After 24 h exposure, CSC decreased dose-dependently cell viability accompanying an increase in intracellular ROS and NO level. CSC structurally or functionally damaged organelles including mitochondria, ER and lysosome and enhanced the expression of proteins related to apoptosis, ER stress and DNA damage accompanying an elevated proportion of annexin V-bound cells. Meanwhile, the expression of certain proteins related to mitochondrial dynamics (OPA1 and DRP1) and autophagy (ATG5) did not overall show significant dose-dependent change in cells exposed to CSC. More importantly, conversion of LC3-I to LC3B-II, a representative marker for autophagy, was also unclear. Considering that intracellular organelles work together in harmony to perform defense mechanism against foreign bodies, we investigated changes in immune response following CSC exposure. The level of IFN-γ and MIP-1α was elevated in CSC-exposed cells, whereas the MCP-1α level decreased. The expression of chemokine receptors (CD195 and CXCR2) and an adhesion molecule (CD54) increased by CSC treatment, the expression of certain antigen presentation-related proteins (MHC class II, CD40, and CD80) were also enhanced. Meanwhile, the expression of CD86, a co-stimulatory molecule for antigen presentation, dose-dependently decreased. In conclusion, we suggest that CSC may induce apoptotic cell death and disturbance in host defense mechanisms by impairing function of cellular components.


Subject(s)
Macrophages, Alveolar/drug effects , Organelles/drug effects , Smoke/adverse effects , Tobacco Products , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cell Line , Cytokines/metabolism , DNA Damage , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/ultrastructure , Mice , Microscopy, Electron, Transmission , Nitric Oxide/metabolism , Organelles/ultrastructure , Reactive Oxygen Species/metabolism , Receptors, CCR5/metabolism , Receptors, Interleukin-8B/metabolism
18.
Mol Immunol ; 101: 130-139, 2018 09.
Article in English | MEDLINE | ID: mdl-29935435

ABSTRACT

Interleukin-1ß (IL-1ß) is a critical inflammatory regulator in response to Mycoplasma hyopneumoniae infection. However, the mechanism involved in the secretion of IL-1ß during Mycoplasma hyopneumoniae infection is unclear. In this study, we demonstrated that Mycoplasma hyopneumoniae infection increased the secretion of mature-IL-1ß (m-IL-1ß), but not pro-IL-1ß, in porcine alveolar macrophages. Moreover, Mycoplasma hyopneumoniae infection promoted the generation of autophagosomes, which attributed to the unconventional secretion of m-IL-1ß. Further results revealed that Hsp90 was required for the entry of m-IL-1ß into autophagosomes during Mycoplasma hyopneumoniae infection. The fusion of m-IL-1ß-containing autophagosome and plasma membranes was regulated by Sec22b and independent of lysosomal dysfunction. In conclusion, we provide evidence that Hsp90/Sec22b promotes the unconventional secretion of IL-1ß through an autophagosomal carrier during Mycoplasma hyopneumoniae infection. The elucidation of the molecular and cellular machinery in Mycoplasma hyopneumoniae infected mammalian cells in this study suggests avenues for further study and applications and paves the way for novel therapeutic strategies to prevent tissue damage in mycoplasma-associated diseases.


Subject(s)
Autophagosomes/metabolism , HSP90 Heat-Shock Proteins/metabolism , Interleukin-1beta/metabolism , Macrophages, Alveolar/microbiology , Mycoplasma hyopneumoniae/physiology , Pneumonia of Swine, Mycoplasmal/metabolism , Pneumonia of Swine, Mycoplasmal/microbiology , R-SNARE Proteins/metabolism , Animals , Cell Membrane/metabolism , Lysosomes/metabolism , Macrophages, Alveolar/ultrastructure , Mycoplasma hyopneumoniae/ultrastructure , Swine
19.
Pathog Dis ; 76(3)2018 04 01.
Article in English | MEDLINE | ID: mdl-29718182

ABSTRACT

The pathogenesis of Mycobacterium tuberculosis (Mtb) is intrinsically linked to its intimate and enduring interaction with its host, and understanding Mtb-host interactions at a molecular level is critical to attempts to decrease the significant burden of tuberculosis disease. The marked heterogeneity that exists in lesion progression and outcome during Mtb infection necessitates the development of methods that enable in situ analyses of Mtb biology and host response within the spatial context of tissue structure. Fluorescent reporter Mtb strains have thus come to the forefront as an approach with broad utility for the study of the Mtb-host interface, enabling visualization of the bacteria during infection, and contributing to the discovery of several facets such as non-uniformity in microenvironments and Mtb physiology in vivo, and their relation to the host immune response or therapeutic intervention. We review here the different types of fluorescent reporters and ways in which they have been utilized in Mtb studies, and expand on how they may further be exploited in combination with novel imaging and other methodologies to illuminate key aspects of Mtb-host interactions.


Subject(s)
Genes, Reporter , Host-Pathogen Interactions , Macrophages, Alveolar/microbiology , Mycobacterium tuberculosis/metabolism , Mycobacterium/metabolism , Tuberculosis, Pulmonary/microbiology , Animals , Antitubercular Agents/therapeutic use , Cell Tracking/methods , Gene Expression , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Lung/microbiology , Lung/pathology , Lung/ultrastructure , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/ultrastructure , Mice , Mycobacterium/drug effects , Mycobacterium/genetics , Mycobacterium/ultrastructure , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/ultrastructure , Optical Imaging/methods , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/pathology , Red Fluorescent Protein
20.
Med Mycol J ; 59(1): E1-E6, 2018.
Article in Japanese | MEDLINE | ID: mdl-29491337

ABSTRACT

This article presents the ultrastructural patterns of interactions between the murine lung macrophages and cells of low- (RKPGY-881, -1165, -1178) and high-virulence (RKPGY-1090, -1095, -1106) strains of Cryptococcus neoformans at the seventh post-experimental day. It was found that if macrophages ingest living yeast cells, the latter can: 1) become completely free from polysaccharide capsules, after that their contents undergo lysis, and cell wall debris are extruded from the macrophage (first scenario); 2) become partly free from their capsules, destroy the phagosomal plasma membrane and induce destructive processes inside the macrophage causing their death (second scenario); or 3) not lose their capsules and localize inside macrophage in latent state (third scenario). Macrophages can also ingest senescent and dead C. neoformans cells surrounded by capsules that are lost at the ingesting and phagosome stages (fourth scenario). The study revealed the dependence of cell-mediated immunity on the stage of development of ingested C. neoformans yeast cells. Here we describe a new mechanism of capsular polysaccharide elimination of C. neoformans yeast cells by murine macrophages.


Subject(s)
Cryptococcus neoformans/immunology , Cryptococcus neoformans/ultrastructure , Macrophages, Alveolar/immunology , Macrophages, Alveolar/ultrastructure , Phagocytosis , Animals , Cryptococcus neoformans/metabolism , Cryptococcus neoformans/pathogenicity , Fungal Capsules/metabolism , Fungal Capsules/ultrastructure , Fungal Polysaccharides/metabolism , Immunity, Cellular/immunology , Male , Mice , Phagosomes , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...