Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
Nutrients ; 16(11)2024 May 24.
Article in English | MEDLINE | ID: mdl-38892531

ABSTRACT

Propolis has potential anti-inflammatory properties, but little is known about its efficacy against inflammatory reactions caused by drug-resistant bacteria, and the difference in efficacy between propolis and tree gum is also unclear. Here, an in vivo study was performed to study the effects of ethanol extract from poplar propolis (EEP) and poplar tree gum (EEG) against heat-inactivated methicillin-resistant Staphylococcus aureus (MRSA)-induced acute lung injury (ALI) in mice. Pre-treatment with EEP and EEG (100 mg/kg, p.o.) resulted in significant protective effects on ALI in mice, and EEP exerted stronger activity to alleviate lung tissue lesions and ALI scores compared with that of EEG. Furthermore, EEP significantly suppressed the levels of pro-inflammatory mediators in the lung, including TNF-α, IL-1ß, IL-6, and IFN-γ. Gut microbiota analysis revealed that both EEP and EEG could modulate the composition of the gut microbiota, enhance the abundance of beneficial microbiota and reduce the harmful ones, and partly restore the levels of short-chain fatty acids. EEP could modulate more serum metabolites and showed a more robust correlation between serum metabolites and gut microbiota. Overall, these results support the anti-inflammatory effects of propolis in the treatment of ALI, and the necessity of the quality control of propolis.


Subject(s)
Acute Lung Injury , Gastrointestinal Microbiome , Inflammation Mediators , Methicillin-Resistant Staphylococcus aureus , Propolis , Propolis/pharmacology , Animals , Methicillin-Resistant Staphylococcus aureus/drug effects , Acute Lung Injury/microbiology , Acute Lung Injury/drug therapy , Gastrointestinal Microbiome/drug effects , Mice , Male , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Anti-Inflammatory Agents/pharmacology , Staphylococcal Infections/drug therapy , Cytokines/blood , Cytokines/metabolism , Hot Temperature , Disease Models, Animal
2.
Zhongguo Zhong Yao Za Zhi ; 49(7): 1915-1923, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38812204

ABSTRACT

This study aims to elucidate the therapeutic effect and mechanism of Jingfang Granules on acute lung injury, and to investigate the regulatory effect of Jingfang Granules on the metabolic disorders of endogenous metabolites in feces and the homeostasis of intestinal microbiota in acute lung injury, mice were randomly divided into a sham group, a model group, and a Jingfang Granules group. After modeling, the mice were continuously administered for 6 days. Using ultra-high performance liquid chromatography quadrupole/electrostatic field orbital trap high-resolution mass spectrometry(UHPLC-HESI-QE-Orbitrap-MS/MS) metabolomics technology and 16S rRNA high-throughput sequencing technology, changes in endogenous small molecule substances and gut microbiota in mouse intestines were determined, and potential biomarkers were identified. The results showed that Jingfang Granules can regulate 11 biomarkers, including L-glutamic acid, succinic acid, arachidonic acid, linoleic acid, linolenic acid, phenylalanine, sphingosine, 2-hydroxy-2-methyl butyric acid, pyruvate, tryptophan, and palmitic acid. Metabolic pathway analysis was conducted on these 11 biomarkers using the online software MetaboAnalyst, identifying potential major metabolic pathways. Among them, a total of 10 metabolic pathways are closely related to the treatment of acute lung injury with Jingfang Granules, including alanine, aspartate and glutamate metabolism, aminoacyl-tRNA biosynthesis, citrate cycle(TCA cycle), alyoxylate and dicarboxylate metabolism, arginine and proline metabolism, linoleic acid metabolism and linolenic acid metabolism, nitrogen metabolism, D-glutamine and D-gluta-matemetabolism, phenylalanine, tyrosine and tryptophan biosynthesis, phenylalanine metabolism. The results of gut microbiota showed significant differences in bacteria, mainly including Bacteroides, Akkermansia, Lachnospiraceae_NK4A136_group, Lachnochlostridium, and Klebsiella. Spearman analysis confirms that Akkermansia and Lachnospiraceae_NK4A136_group is a significant positive correlation between the abundance of succinic acid, arachidonic acid, linolenic acid, linoleic acid, butyric acid, and pyruvate in the group; Bacteroides, Klebsiella, Lachnochlostrium are significantly positively correlated with the abundance of L-glutamic acid, phenylalanine, and sphingosine. The above results indicate that the therapeutic effect of Jingfang Granules on acute lung injury is achieved by improving the imbalance of gut microbiota in mice with acute lung injury, balancing the metabolism of alanine, biosynthesis of aminoacyl tRNA, aspartic acid, glutamate, tricarboxylic acid cycle, biosynthesis of phenylalanine, tyrosine, tryptophan, and metabolism of linoleic acid.


Subject(s)
Acute Lung Injury , Drugs, Chinese Herbal , Feces , Gastrointestinal Microbiome , Metabolomics , Animals , Mice , Gastrointestinal Microbiome/drug effects , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/microbiology , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/administration & dosage , Male , Feces/microbiology , Feces/chemistry , Humans , Chromatography, High Pressure Liquid
3.
Anaerobe ; 87: 102854, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38614288

ABSTRACT

OBJECTIVES: Acute lung injury is a critical complication of severe acute pancreatitis (SAP). The gut microbiota and its metabolites play an important role in SAP development and may provide new targets for AP-associated lung injury. Based on the ability to reverse AP injury, we proposed that Clostridium butyricum may reduce the potential for AP-associated lung injury by modulating with intestinal microbiota and related metabolic pathways. METHODS: An AP disease model was established in mice and treated with C. butyricum. The structure and composition of the intestinal microbiota in mouse feces were analyzed by 16 S rRNA gene sequencing. Non-targeted metabolite analysis was used to quantify the microbiota derivatives. The histopathology of mouse pancreas and lung tissues was examined using hematoxylin-eosin staining. Pancreatic and lung tissues from mice were stained with immunohistochemistry and protein immunoblotting to detect inflammatory factors IL-6, IL-1ß, and MCP-1. RESULTS: C. butyricum ameliorated the dysregulation of microbiota diversity in a model of AP combined with lung injury and affected fatty acid metabolism by lowering triglyceride levels, which were closely related to the alteration in the relative abundance of Erysipelatoclostridium and Akkermansia. In addition, C. butyricum treatment attenuated pathological damage in the pancreatic and lung tissues and significantly suppressed the expression of inflammatory factors in mice. CONCLUSIONS: C. butyricum may alleviate lung injury associated with AP by interfering with the relevant intestinal microbiota and modulating relevant metabolic pathways.


Subject(s)
Clostridium butyricum , Disease Models, Animal , Gastrointestinal Microbiome , Metabolomics , Pancreatitis , RNA, Ribosomal, 16S , Animals , RNA, Ribosomal, 16S/genetics , Mice , Pancreatitis/microbiology , Pancreatitis/metabolism , Pancreatitis/pathology , Metabolomics/methods , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Probiotics/administration & dosage , Male , Feces/microbiology , Pancreas/pathology , Pancreas/microbiology , Lung/microbiology , Lung/pathology
4.
Int J Mol Sci ; 25(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38673868

ABSTRACT

This study aimed to investigate the preventive effects of the total polyphenols from Nymphaea candida (NCTP) on LPS-induced septic acute lung injury (ALI) in mice and its mechanisms. NCTP could significantly ameliorate LPS-induced lung tissue pathological injury in mice as well as lung wet/dry ratio and MPO activities (p < 0.05). NCTP could significantly decrease the blood leukocyte, neutrophil, monocyte, basophil, and eosinophil amounts and LPS contents in ALI mice compared with the model group (p < 0.05), improving lymphocyte amounts (p < 0.05). Moreover, compared with the model group, NCTP could decrease lung tissue TNF-α, IL-6, and IL-1ß levels (p < 0.05) and downregulate the protein expression of TLR4, MyD88, TRAF6, IKKß, IκB-α, p-IκB-α, NF-κB p65, p-NF-κB p65, NLRP3, ASC, and Caspase1 in lung tissues (p < 0.05). Furthermore, NCTP could inhibit ileum histopathological injuries, restoring the ileum tight junctions by increasing the expression of ZO-1 and occludin. Simultaneously, NCTP could reverse the gut microbiota disorder, restore the diversity of gut microbiota, increase the relative abundance of Clostridiales and Lachnospiraceae, and enhance the content of SCFAs (acetic acid, propionic acid, and butyric acid) in feces. These results suggested that NCTP has preventive effects on septic ALI, and its mechanism is related to the regulation of gut microbiota, SCFA metabolism, and the TLR-4/NF-κB and NLRP3 pathways.


Subject(s)
Acute Lung Injury , Gastrointestinal Microbiome , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Polyphenols , Sepsis , Signal Transduction , Toll-Like Receptor 4 , Animals , Acute Lung Injury/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/prevention & control , Acute Lung Injury/microbiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Toll-Like Receptor 4/metabolism , Gastrointestinal Microbiome/drug effects , Mice , NF-kappa B/metabolism , Polyphenols/pharmacology , Sepsis/complications , Sepsis/metabolism , Signal Transduction/drug effects , Male , Lipopolysaccharides
5.
ACS Appl Mater Interfaces ; 15(1): 494-510, 2023 Jan 11.
Article in English | MEDLINE | ID: mdl-36577517

ABSTRACT

Targeting the limitation of antimicrobial peptides (AMPs) application in vivo, self-assembled AMPs library with specific nanostructures is expected to gradually overtake monomer AMPs libraries in the future. Peptide polymers are fascinating self-assembling nanoscale structures that have great advantage in biomedical applications because of their satisfactory biocompatibility and versatile properties. Herein, we describe a strategy for inducing the self-assembly of T9W into nanostructured antimicrobial micelles with evidently improved pharmacological properties, that is, PEGylation at the C-terminal of T9W (CT9W1000), an antibacterial biomaterial that self-assembles in aqueous media without exogenous excipients, has been developed. Compared with parental molecular, the CT9W1000 is more effective against Pseudomonas aeruginosa, and its antibacterial spectrum had also been broadened. Additionally, CT9W1000 micelles had higher stability under salt ion, serum, and acid-base environments. Importantly, the self-assembled structure is highly resistant to trypsin degradation, probably allowing T9W to be applied in clinical settings in the future. Mechanistically, by acting on membranes and through supplementary bactericidal mechanisms, CT9W1000 micelles contribute to the antibacterial process. Collectively, CT9W1000 micelles exhibited good biocompatibility in vitro and in vivo, resulting in highly effective treatment in a mouse acute lung injury model induced by P. aeruginosa PAO1 without drug resistance. These advances may profoundly accelerate the clinical transformation of T9W and promote the development of a combination of peptide-based antibiotics and PEGylated nanotechnology.


Subject(s)
Acute Lung Injury , Antimicrobial Peptides , Micelles , Pseudomonas aeruginosa , Animals , Mice , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Cationic Peptides/chemistry , Disease Models, Animal , Microbial Sensitivity Tests , Trypsin/metabolism , Acute Lung Injury/drug therapy , Acute Lung Injury/etiology , Acute Lung Injury/microbiology , Nanostructures/chemistry , Pseudomonas Infections/complications , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Drug Resistance, Bacterial
6.
Am J Physiol Lung Cell Mol Physiol ; 322(1): L149-L161, 2022 01 01.
Article in English | MEDLINE | ID: mdl-35015568

ABSTRACT

Disruption of the lung endothelial barrier is a hallmark of acute respiratory distress syndrome (ARDS), for which no effective pharmacologic treatments exist. Prior work has demonstrated that FTY720 S-phosphonate (Tys), an analog of sphingosine-1-phosphate (S1P) and FTY720, exhibits potent endothelial cell (EC) barrier protective properties. In this study, we investigated the in vitro and in vivo efficacy of Tys against methicillin-resistant Staphylococcus aureus (MRSA), a frequent bacterial cause of ARDS. Tys-protected human lung EC from barrier disruption induced by heat-killed MRSA (HK-MRSA) or staphylococcal α-toxin and attenuated MRSA-induced cytoskeletal changes associated with barrier disruption, including actin stress fiber formation and loss of peripheral VE-cadherin and cortactin. Tys-inhibited Rho and myosin light chain (MLC) activation after MRSA and blocked MRSA-induced NF-κB activation and release of the proinflammatory cytokines, IL-6 and IL-8. In vivo, intratracheal administration of live MRSA in mice caused significant vascular leakage and leukocyte infiltration into the alveolar space. Pre- or posttreatment with Tys attenuated MRSA-induced lung permeability and levels of alveolar neutrophils. Posttreatment with Tys significantly reduced levels of bronchoalveolar lavage (BAL) VCAM-1 and plasma IL-6 and KC induced by MRSA. Dynamic intravital imaging of mouse lungs demonstrated Tys attenuation of HK-MRSA-induced interstitial edema and neutrophil infiltration into lung tissue. Tys did not directly inhibit MRSA growth or viability in vitro. In conclusion, Tys inhibits lung EC barrier disruption and proinflammatory signaling induced by MRSA in vitro and attenuates acute lung injury induced by MRSA in vivo. These results support the potential utility of Tys as a novel ARDS therapeutic strategy.


Subject(s)
Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Cell Membrane Permeability , Endothelial Cells/microbiology , Fingolimod Hydrochloride/analogs & derivatives , Methicillin-Resistant Staphylococcus aureus/physiology , Organophosphonates/pharmacology , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Cell Membrane Permeability/drug effects , Cytoprotection/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Enzyme Activation/drug effects , Fingolimod Hydrochloride/pharmacology , Humans , Inflammation/pathology , Mice , Myosin Light Chains/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , rhoA GTP-Binding Protein/metabolism
7.
Physiol Rep ; 9(22): e15116, 2021 11.
Article in English | MEDLINE | ID: mdl-34822216

ABSTRACT

Obesity alters the risks and outcomes of inflammatory lung diseases. It is important to accurately recapitulate the obese state in animal models to understand these effects on the pathogenesis of disease. Diet-induced obesity is a commonly used model of obesity, but when applied to other disease models like acute respiratory distress syndrome, pneumonia, and asthma, it yields widely divergent. We hypothesized high-fat chow storage conditions would affect lipid oxidation and inflammatory response in the lungs of lipopolysaccharide (LPS)-challenged mice. For 6 weeks, C57BL/6crl mice were fed either a 10% (low-fat diet, LFD) or 60% (high-fat diet, HFD) stored at room temperature (RT, 23°C) for up to 7, 14, 21, or 42 days. Mice were treated with nebulized LPS to induce lung inflammation, and neutrophil levels in bronchoalveolar lavage were determined 24 h later. Lipid oxidation (malondialdehyde, MDA) was assayed by thiobarbituric acid reactive substances in chow and mouse plasma. Concentrations of MDA in chow and plasma rose in proportion to the duration of RT chow storage. Mice fed a HFD stored <2 weeks at RT had an attenuated response 24 h after LPS compared with mice fed an LFD. This effect was reversed after 2 weeks of chow storage at RT. Chow stored above freezing underwent lipid oxidation associated with significant alterations in the LPS-induced pulmonary inflammatory response. Our data show that storage conditions affect lipid peroxidation, which in turn affects pulmonary inflammatory responses in a mouse model of disease. It also suggests changes in the microbiome, although not significantly different suggests decreased variety and richness of bacteria in the gut, a large aspect of the immune system. Dietary composition and storage of chow may also affect pulmonary inflammation and the gut microbiome in humans.


Subject(s)
Acute Lung Injury/metabolism , Animal Feed , Diet, High-Fat , Food Storage , Inflammation/metabolism , Malondialdehyde/metabolism , Obesity/metabolism , Pneumonia/metabolism , Temperature , Acute Lung Injury/chemically induced , Acute Lung Injury/microbiology , Animals , Diet, Fat-Restricted , Disease Models, Animal , Gastrointestinal Microbiome , Inflammation/microbiology , Lipid Metabolism , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Obesity/microbiology , Pneumonia/chemically induced , Pneumonia/microbiology
8.
Amino Acids ; 53(10): 1609-1622, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34524541

ABSTRACT

Escherichia coli infections can result in lung injury, which may be closely linked to the induction of interferon secretion. The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway is one of most important pathways that regulate interferon production. Thus, the present study aimed to dissect whether E. coli infections can regulate interferon production and the underlying mechanisms. For this aim, two lung cell lines, a human bronchial epithelial cell line transformed with Ad12-SV40 2B (BEAS-2b) and a human fetal lung fibroblast (HFL1) cell line, were used. The effects of E. coli infections on interferon production were studied using qRT-PCR, Western blot, and siRNA knockdown assays. E. coli infections remarkably promoted the expression levels of IFN-α, IFN-ß, and ISGs. Major components of the JAK/STAT pathway, including JAK1, STAT1, and STAT2, were demonstrated to be regulated by E. coli infections. Importantly, knockdown of JAK1, STAT1, and STAT2 abolished the induction of IFN-α, IFN-ß, and ISGs by E. coli. Therefore, experiments in the present study demonstrated that E. coli infections remarkably promoted interferon production in lung cells, which was closely regulated by the JAK/STAT signaling pathway. The findings in the present study are useful for further understanding the pathogenesis of E. coli infections in the lung and finding novel therapies to treat E. coli-induced lung injury.


Subject(s)
Acute Lung Injury/microbiology , Escherichia coli Infections/metabolism , Interferon-alpha/metabolism , Interferon-beta/metabolism , Acute Lung Injury/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Line , Escherichia coli Infections/genetics , Exoribonucleases/genetics , Gene Expression Regulation , Humans , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Lung/cytology , Lung/microbiology , Myxovirus Resistance Proteins/genetics , RNA-Binding Proteins/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Signal Transduction
9.
Am J Physiol Lung Cell Mol Physiol ; 321(5): L827-L836, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34524905

ABSTRACT

We previously reported that extracellular vesicles (EVs) released during Escherichia coli (E. coli) bacterial pneumonia were inflammatory, and administration of high molecular weight hyaluronic acid (HMW HA) suppressed several indices of acute lung injury (ALI) from E. coli pneumonia by binding to these inflammatory EVs. The current study was undertaken to study the therapeutic effects of HMW HA in ex vivo perfused human lungs injured with Pseudomonas aeruginosa (PA)103 bacterial pneumonia. For lungs with baseline alveolar fluid clearance (AFC) <10%/h, HMW HA 1 or 2 mg was injected intravenously after 1 h (n = 4-9), and EVs released during PA pneumonia were collected from the perfusate over 6 h. For lungs with baseline AFC > 10%/h, HMW HA 2 mg was injected intravenously after 1 h (n = 6). In vitro experiments were conducted to evaluate the effects of HA on inflammation and bacterial phagocytosis. For lungs with AFC < 10%/h, administration of HMW HA intravenously significantly restored AFC and numerically decreased protein permeability and alveolar inflammation from PA103 pneumonia but had no effect on bacterial counts at 6 h. However, HMW HA improved bacterial phagocytosis by human monocytes and neutrophils and suppressed the inflammatory properties of EVs released during pneumonia on monocytes. For lungs with AFC > 10%/h, administration of HMW HA intravenously improved AFC from PA103 pneumonia but had no significant effects on protein permeability, inflammation, or bacterial counts. In the presence of impaired alveolar epithelial transport capacity, administration of HMW HA improved the resolution of pulmonary edema from Pseudomonas PA103 bacterial pneumonia.


Subject(s)
Acute Lung Injury/drug therapy , Hyaluronic Acid/pharmacology , Pneumonia, Bacterial/drug therapy , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Pulmonary Edema/drug therapy , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Adult , Extracellular Vesicles/pathology , Female , Humans , Lung/drug effects , Lung/microbiology , Lung/pathology , Male , Middle Aged , Monocytes/immunology , Neutrophils/immunology , Organ Culture Techniques , Phagocytosis/drug effects , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , Pulmonary Edema/microbiology , Pulmonary Edema/pathology , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/microbiology , Respiratory Distress Syndrome/pathology
10.
Front Immunol ; 12: 693874, 2021.
Article in English | MEDLINE | ID: mdl-34349759

ABSTRACT

Background: The mechanisms by which moderate tidal volume ventilation (MTV) exacerbates preexisting lung injury are unclear. We hypothesized that systemic endotoxemia via the gut-lung axis would lead to non-canonical and canonical inflammasome activation and pyroptosis in a two-hit model involving polyinosinic-polycytidylic acid (Poly(I:C)), a synthetic analog of dsRNA and MTV and that this would associate with acute lung injury (ALI). Methods: Anesthetized mice were administered Poly(I:C) intratracheally and then 6 h later, they were mechanically ventilated for 4 h with otherwise non-injurious MTV (10ml/kg). Changes in intestinal and alveolar capillary permeability were measured. Further documentation of ALI was assessed by evans blue albumin permeability, protein and IL-1 family concentration in bronchoalveolar lavage fluid (BALF) or plasma, and histopathology in cohorts of wildtype (WT), whole body genetically ablated caspase-11 (caspase-11-/-), caspase-1/caspase-11 double knockout (caspase-1/11-/-), gasdermin D (GSDMD)-/-, nucleotide-binding domain leucine-rich repeat-containing protein 3 (NLRP3)-/- and advanced glycosylation end product-specific receptor (RAGE) -/- mice. Results: Non-injurious MTV exacerbated the mild lung injury associated with Poly(I:C) administration. This included the disruption of alveolar-capillary barrier and increased levels of interleukin (IL)-6, high mobility group proteins 1 (HMGB-1), IL-1ß in BALF and IL-18 in plasma. Combined (Poly(I:C)-MTV) injury was associated with increase in gastrointestinal permeability and endotoxin in plasma and BALF. Poly(I:C)-MTV injury was sensitive to caspase-11 deletion with no further contribution of caspase-1 except for maturation and release of IL-18 (that itself was sensitive to deletion of NLRP3). Combined injury led to large increases in caspase-1 and caspase-11. Genetic ablation of GSDMD attenuated alveolar-capillary disruption and release of cytokines in combined injury model. Conclusions: The previously noted exacerbation of mild Poly(I:C)-induced ALI by otherwise non-injurious MTV is associated with an increase in gut permeability resulting in systemic endotoxemia. The gut-lung axis resulted in activation of pulmonary non-canonical (cytosolic mediated caspase-11 activation) and canonical (caspase-1) inflammasome (NLRP3) mediated ALI in this two-hit model resulting in GSDMD sensitive alveolar capillary barrier disruption, pyroptosis (alveolar macrophages) and cytokine maturation and release (IL-1ß; IL-18). Pharmacologic strategies aimed at disrupting communication between gut and lung, inhibition of inflammasomes or GSDMD in pyroptosis may be useful in ALI.


Subject(s)
Acute Lung Injury/chemically induced , Caspases, Initiator/metabolism , Gastrointestinal Microbiome , Intestines/microbiology , Lung/enzymology , Poly I-C , Respiration, Artificial , Ventilator-Induced Lung Injury/etiology , Acute Lung Injury/enzymology , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Animals , Bacteria/metabolism , Caspases, Initiator/genetics , Disease Models, Animal , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/metabolism , Lung/pathology , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , Pyroptosis , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/microbiology , Ventilator-Induced Lung Injury/pathology
11.
Cells ; 10(7)2021 07 08.
Article in English | MEDLINE | ID: mdl-34359901

ABSTRACT

Lung endothelial dysfunction is a key feature of acute lung injury (ALI) and clinical acute respiratory distress syndrome (ARDS). Previous studies have identified the lipid-generating enzyme, group V phospholipase A2 (gVPLA2), as a mediator of lung endothelial barrier disruption and inflammation. The current study aimed to determine the role of gVPLA2 in mediating lung endothelial responses to methicillin-resistant Staphylococcus aureus (MRSA, USA300 strain), a major cause of ALI/ARDS. In vitro studies assessed the effects of gVPLA2 inhibition on lung endothelial cell (EC) permeability after exposure to heat-killed (HK) MRSA. In vivo studies assessed the effects of intratracheal live or HK-MRSA on multiple indices of ALI in wild-type (WT) and gVPLA2-deficient (KO) mice. In vitro, HK-MRSA increased gVPLA2 expression and permeability in human lung EC. Inhibition of gVPLA2 with either the PLA2 inhibitor, LY311727, or with a specific monoclonal antibody, attenuated the barrier disruption caused by HK-MRSA. LY311727 also reduced HK-MRSA-induced permeability in mouse lung EC isolated from WT but not gVPLA2-KO mice. In vivo, live MRSA caused significantly less ALI in gVPLA2 KO mice compared to WT, findings confirmed by intravital microscopy assessment in HK-MRSA-treated mice. After targeted delivery of gVPLA2 plasmid to lung endothelium using ACE antibody-conjugated liposomes, MRSA-induced ALI was significantly increased in gVPLA2-KO mice, indicating that lung endothelial expression of gVPLA2 is critical in vivo. In summary, these results demonstrate an important role for gVPLA2 in mediating MRSA-induced lung EC permeability and ALI. Thus, gVPLA2 may represent a novel therapeutic target in ALI/ARDS caused by bacterial infection.


Subject(s)
Acute Lung Injury/enzymology , Acute Lung Injury/microbiology , Endothelial Cells/microbiology , Endothelial Cells/pathology , Methicillin-Resistant Staphylococcus aureus/physiology , Phospholipases A2/metabolism , Acute Lung Injury/pathology , Animals , Cell Membrane Permeability/drug effects , Endothelial Cells/drug effects , Indoles/pharmacology , Lung/diagnostic imaging , Lung/microbiology , Lung/pathology , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice, Knockout , Models, Biological , Phospholipases A2/deficiency
12.
FASEB J ; 35(9): e21797, 2021 09.
Article in English | MEDLINE | ID: mdl-34383981

ABSTRACT

Pseudomonas aeruginosa is a frequent cause of hospital-acquired lung infections characterized by hyperinflammation, antibiotic resistance, and high morbidity/mortality. Here, we show that the genetic ablation of one cAMP-phosphodiesterase 4 subtype, PDE4B, is sufficient to protect mice from acute lung injury induced by P aeruginosa infection as it reduces pulmonary and systemic levels of pro-inflammatory cytokines, as well as pulmonary vascular leakage and mortality. Surprisingly, despite dampening immune responses, bacterial clearance in the lungs of PDE4B-KO mice is significantly improved compared to WT controls. In wildtypes, P aeruginosa-infection produces high systemic levels of several cytokines, including TNF-α, IL-1ß, and IL-6, that act as cryogens and render the animals hypothermic. This, in turn, diminishes their ability to clear the bacteria. Ablation of PDE4B curbs both the initial production of acute response cytokines, including TNF-α and IL-1ß, as well as their downstream signaling, specifically the induction of the secondary-response cytokine IL-6. This synergistic action protects PDE4B-KO mice from the deleterious effects of the P aeruginosa-induced cytostorm, while concurrently improving bacterial clearance, rather than being immunosuppressive. These benefits of PDE4B ablation are in contrast to the effects resulting from treatment with PAN-PDE4 inhibitors, which have been shown to increase bacterial burden and dissemination. Thus, PDE4B represents a promising therapeutic target in settings of P aeruginosa lung infections.


Subject(s)
Acute Lung Injury/metabolism , Acute Lung Injury/microbiology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Hypothermia/metabolism , Hypothermia/microbiology , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/pathogenicity , Animals , Cytokines/metabolism , Lung/metabolism , Lung/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphodiesterase 4 Inhibitors/pharmacology , Pseudomonas Infections/microbiology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism
13.
J Trauma Acute Care Surg ; 90(6): 924-934, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34016916

ABSTRACT

BACKGROUND: Forty percent of critically ill trauma patients will develop an infectious complication. Pneumonia is the most common cause of death of trauma patients surviving their initial insult. We previously demonstrated that polytrauma (PT), defined as two or more severe injuries in at least two areas of the body, induces emergency hematopoiesis characterized by accelerated myelopoiesis in the bone marrow and increased myeloid cell frequency in the peripheral tissues. We hypothesized that PT alone induces priming of neutrophils, resulting in hyperactivation upon secondary exposure to bacteria and causing acute lung injury and increased susceptibility to secondary exposure to Pseudomonas aeruginosa pneumonia. METHODS: C57BL/6 mice were subjected to PT consisting of a lower extremity pseudofracture, liver crush injury, and 15% blood-volume hemorrhage. Pneumonia was induced by intratracheal injection of 5 × 106 CFU live P. aeruginosa or 1 × 107 of heat-killed P. aeruginosa (HKPA). For reactive oxygen species (ROS), studies polymorphonuclear neutrophils (PMNs) were isolated by immunomagnetic bead negative selection and stimulated ex-vivo with HKPA. Reactive oxygen species production was measured by immunofluorescence. For histology, lung sections were stained by hematoxylin-eosin and analyzed by a blinded grader. RESULTS: Polytrauma induced persistent changes in immune function at baseline and to secondary infection. Pneumonia after injury resulted in increased mortality (60% vs. 5% p < 0.01). Blood neutrophils from PT mice had higher resting (unstimulated) ROS production than in naive animals (p < 0.02) demonstrating priming of the neutrophils following PT. After intratracheal HKPA injection, bronchoalveolar lavage PMNs from injured mice had higher ROS production compared with naive mice (p < 0.01), demonstrating an overexuberant immunopathologic response of neutrophils following PT. CONCLUSION: Polytrauma primes neutrophils and causes immunopathologic PMN ROS production, increased lung injury and susceptibility to secondary bacterial pneumonia. These results suggest that trauma-induced immune dysfunction can cause immunopathologic response to secondary infection and suggests neutrophil-mediated pulmonary damage as a therapeutic target for posttrauma pneumonia.


Subject(s)
Acute Lung Injury/immunology , Multiple Trauma/complications , Neutrophils/immunology , Pneumonia, Bacterial/immunology , Pseudomonas Infections/immunology , Acute Lung Injury/blood , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Animals , Disease Models, Animal , Humans , Lung/immunology , Lung/microbiology , Lung/pathology , Male , Mice , Multiple Trauma/blood , Multiple Trauma/diagnosis , Multiple Trauma/immunology , Neutrophils/metabolism , Pneumonia, Bacterial/blood , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , Pseudomonas Infections/blood , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/immunology , Reactive Oxygen Species/metabolism , Trauma Severity Indices
14.
Am J Respir Cell Mol Biol ; 64(5): 569-578, 2021 05.
Article in English | MEDLINE | ID: mdl-33571420

ABSTRACT

Pneumonia-induced lung injury and acute respiratory distress syndrome can develop because of an inappropriate inflammatory response to acute infections, leading to a compromised alveolar barrier. Recent work suggests that hospitalized patients with allergies/asthma are less likely to die of pulmonary infections and that there is a correlation between survival from acute respiratory distress syndrome and higher eosinophil counts; thus, we hypothesized that eosinophils associated with a type 2 immune response may protect against pneumonia-induced acute lung injury. To test this hypothesis, mice were treated with the type 2-initiating cytokine IL-33 intratracheally 3 days before induction of pneumonia with airway administration of a lethal dose of Staphylococcus aureus. Interestingly, IL-33 pretreatment promoted survival by inhibiting acute lung injury: amount of BAL fluid proinflammatory cytokines and pulmonary edema were both reduced, with an associated increase in oxygen saturation. Pulmonary neutrophilia was also reduced, whereas eosinophilia was strongly increased. This eosinophilia was key to protection; eosinophil reduction eliminated both IL-33-mediated protection against mortality and inhibition of neutrophilia and pulmonary edema. Together, these data reveal a novel role for eosinophils in protection against lung injury and suggest that modulation of pulmonary type 2 immunity may represent a novel therapeutic strategy.


Subject(s)
Acute Lung Injury/immunology , Eosinophils/immunology , Interleukin-33/immunology , Pneumonia, Staphylococcal/immunology , Pulmonary Edema/immunology , Respiratory Distress Syndrome/immunology , Staphylococcus aureus/pathogenicity , Acute Lung Injury/etiology , Acute Lung Injury/microbiology , Acute Lung Injury/prevention & control , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Diphtheria Toxin/pharmacology , Disease Models, Animal , Eosinophils/drug effects , Female , Gene Expression , Humans , Interleukin-33/genetics , Interleukin-33/pharmacology , Interleukin-5/deficiency , Interleukin-5/genetics , Interleukin-5/immunology , Leukocyte Count , Leukocyte Reduction Procedures , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/drug effects , Neutrophils/immunology , Pneumonia, Staphylococcal/complications , Pneumonia, Staphylococcal/microbiology , Pneumonia, Staphylococcal/mortality , Pulmonary Edema/complications , Pulmonary Edema/microbiology , Pulmonary Edema/mortality , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/microbiology , Respiratory Distress Syndrome/prevention & control , Staphylococcus aureus/immunology , Survival Analysis
15.
Biomed Res Int ; 2021: 1419631, 2021.
Article in English | MEDLINE | ID: mdl-33506010

ABSTRACT

Lung injury was the common and serious complication of sepsis, a systemic inflammatory response syndrome caused by severe infections. Chinese medicine had unique advantages in attenuating inflammatory response, such as Zuojinfang (ZJF). ZJF was a classical compound herb formula composed of Coptidis Rhizoma and Euodiae Fructus in a ratio of 6 : 1. In this paper, 15 ingredients in ZJF were identified and 8 of them absorbed into rat's serum were quantified by HPLC-MS/MS. Subsequently, sepsis-induced lung injury model was replicated in rats by cecal ligation and puncture. 60 SD rats were randomly divided into 6 groups (n = 10): control group (CON), sham group (Sham), model group (MOD), ZJF low-dose group (ZJF-L), ZJF high-dose group (ZJF-H), and prednisolone group (PNSL). Within the next 24 h, the levels of inflammatory factors, correlation between active ingredients and inflammatory cytokines, the pathological changes of lung tissue, and protein expression of the JAK1/STAT3 signaling pathways were analyzed one by one. Finally, the concentration order of components absorbed in rat serum was berberine > palmatine > jatrorrhizine > coptisine > evodin > chlorogenic acid > evodiamine. Compared with the MOD group, the TNF-α, IL-6, and IFN-γ in the ZJF-H group were significantly reduced (p < 0.05). Moreover, the TNF-α decreased significantly accompanied by the increase of berberine, chlorogenic acid, jatrorrhizine, palmatine, evodin, and evodiamine in serum (negative correlation, p < 0.05). Compared with the MOD, the area of lung injury, the expressions of JAK1, p-JAK1, STAT3, and p-STAT3 were significantly decreased under the treatment of ZJF (p < 0.05). Therefore, downregulating the JAK1/STAT3 signaling pathways was a potential avenue of ZJF in reversing lung injury induced by sepsis.


Subject(s)
Acute Lung Injury/drug therapy , Drugs, Chinese Herbal/pharmacology , Janus Kinase 1/metabolism , STAT3 Transcription Factor/metabolism , Sepsis/pathology , Acute Lung Injury/metabolism , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Animals , Disease Models, Animal , Down-Regulation/drug effects , Janus Kinase 1/genetics , Male , Protective Agents/pharmacology , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/genetics , Sepsis/drug therapy , Sepsis/metabolism , Signal Transduction
16.
J Biol Chem ; 296: 100239, 2021.
Article in English | MEDLINE | ID: mdl-33372035

ABSTRACT

Proinflammatory cytokines such as IL-6 induce endothelial cell (EC) barrier disruption and trigger an inflammatory response in part by activating the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway. The protein suppressor of cytokine signaling-3 (SOCS3) is a negative regulator of JAK-STAT, but its role in modulation of lung EC barrier dysfunction caused by bacterial pathogens has not been investigated. Using human lung ECs and EC-specific SOCS3 knockout mice, we tested the hypothesis that SOCS3 confers microtubule (MT)-mediated protection against endothelial dysfunction. SOCS3 knockdown in cultured ECs or EC-specific SOCS3 knockout in mice resulted in exacerbated lung injury characterized by increased permeability and inflammation in response to IL-6 or heat-killed Staphylococcus aureus (HKSA). Ectopic expression of SOCS3 attenuated HKSA-induced EC dysfunction, and this effect required assembled MTs. SOCS3 was enriched in the MT fractions, and treatment with HKSA disrupted SOCS3-MT association. We discovered that-in addition to its known partners gp130 and JAK2-SOCS3 interacts with MT plus-end binding proteins CLIP-170 and CLASP2 via its N-terminal domain. The resulting SOCS3-CLIP-170/CLASP2 complex was essential for maximal SOCS3 anti-inflammatory effects. Both IL-6 and HKSA promoted MT disassembly and disrupted SOCS3 interaction with CLIP-170 and CLASP2. Moreover, knockdown of CLIP-170 or CLASP2 impaired SOCS3-JAK2 interaction and abolished the anti-inflammatory effects of SOCS3. Together, these findings demonstrate for the first time an interaction between SOCS3 and CLIP-170/CLASP2 and reveal that this interaction is essential to the protective effects of SOCS3 in lung endothelium.


Subject(s)
Inflammation/genetics , Lung Injury/genetics , Microtubule-Associated Proteins/genetics , Neoplasm Proteins/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Acute Lung Injury/genetics , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Animals , Cytoskeleton/genetics , Endothelial Cells , Endothelium, Vascular/metabolism , Endothelium, Vascular/microbiology , Endothelium, Vascular/pathology , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Intercellular Junctions/genetics , Interleukin-6/genetics , Lung Injury/metabolism , Lung Injury/microbiology , Lung Injury/pathology , Mice , Mice, Knockout , Permeability , Staphylococcus aureus/pathogenicity
17.
Int Immunopharmacol ; 91: 107272, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33360370

ABSTRACT

Acute lung injury (ALI) is a common acute respiratory disease treated in the clinic. Intestinal microflora disorder affect lung diseases through the gut-lung axis. In this study, we explored the regulatory mechanism of the gut flora in the host defense against lipopolysaccharide (LPS)-induced ALI through the TLR4/NF-kB pathway by constructing a gut microflora dysbiosis-model with antibiotic administration and reconstruction of the intestinal microecology. Then, high-throughput sequencing was performed, and the levels of secreted IgA (sIgA), ß-defensins, and Muc2 were measured to assess the gut flora and mucosal barrier. The expression of TLR4, NF-kB, TNF-α, IL-1ß, oxidative stress and the lung wet/dry (W/D) ratio were evaluated to assess lung damage. Hematoxylin and eosin (HE) staining was performed to evaluate the damage to the gut and lung tissues. Accordingly, gut microbiota imbalance may regulate the TLR4/NF-kB signaling pathway in the lung immune system, activating oxidative stress in the lung and mediating lung injury through the regulation of the gut barrier. However, fecal microbiota transplantation (FMT) impairs the activity of the TLR4/NF-kB signaling pathway in the lung and decreases oxidative stress in animals with ALI by restoring the gut microecology. CONCLUSIONS: Our results indicated the protective effect of gut flora in regulating immunity of LPS-induced ALI by modulating the TLR4/NF-kB signaling pathway which may induce inflammation and oxidative stress.


Subject(s)
Acute Lung Injury/prevention & control , Bacteria/metabolism , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Intestines/microbiology , Lung/metabolism , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/microbiology , Animals , Anti-Bacterial Agents/toxicity , Bacteria/drug effects , Bacteria/immunology , Disease Models, Animal , Dysbiosis , Gastrointestinal Microbiome/drug effects , Host-Pathogen Interactions , Inflammation Mediators/metabolism , Intestines/drug effects , Lipopolysaccharides , Lung/immunology , Lung/pathology , Mice, Inbred C57BL , Oxidative Stress , Signal Transduction
18.
Int Immunopharmacol ; 90: 107221, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33293260

ABSTRACT

Salvinorin A (SA), a neoclerodane diterpene, is isolated from the dried leaves ofSalvia divinorum. SA has traditionally been used treatments for chronic pain diseases. Recent research has demonstrated that SA possesses the anti-inflammatory property. The present study aim to explore the effects and potentialmechanisms ofSA in protection against Methicillin Resistant Staphylococcus aureus (MRSA)-induced acute lung injury (ALI). Here, we firstly found that verylowdosesof SA (50 µg/kg) could markedly decrease the infiltration of pulmonary neutrophils, mRNA expression of pro-inflammatory cytokines (TNF-α, IL-1ß and IL-6) and then attenuated ALI cause by MRSA infection in mice. In vitro findings revealed that SA attenuated lipoteichoicacid-induced apoptosis, inflammation and oxidative stress in RAW264.7 cells. Mechanism research revealed that SA increased both mRNA levels and protein levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and up-regulated mRNA expression of its downstream genes (HO-1, Gclm, Trx-1, SOD1 and SOD2). Additionally, Nrf2 knockout mice abolished the inhibitory effect of SA on neutrophil accumulation and oxidative stress in MRSA-induced ALI. In conclusion, SA attenuates MRSA-induced ALI via Nrf2 signaling pathways.


Subject(s)
Acute Lung Injury/prevention & control , Anti-Inflammatory Agents/pharmacology , Diterpenes, Clerodane/pharmacology , Lung/drug effects , Methicillin-Resistant Staphylococcus aureus/pathogenicity , NF-E2-Related Factor 2/metabolism , Pneumonia, Staphylococcal/prevention & control , Acute Lung Injury/metabolism , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Animals , Cytokines/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Lung/metabolism , Lung/microbiology , Lung/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/microbiology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , Neutrophil Infiltration/drug effects , Oxidative Stress/drug effects , Pneumonia, Staphylococcal/metabolism , Pneumonia, Staphylococcal/microbiology , Pneumonia, Staphylococcal/pathology , RAW 264.7 Cells , Signal Transduction
19.
Int J Mol Sci ; 21(15)2020 Jul 28.
Article in English | MEDLINE | ID: mdl-32731491

ABSTRACT

Pseudomonas aeruginosa is an important opportunistic pathogen responsible for the cause of acute lung injury and acute respiratory distress syndrome. P. aeruginosa isthe leading species isolated from patients with nosocomial infection and is detected in almost all the patients with long term ventilation in critical care units. P. aeruginosa infection is also the leading cause of deleterious chronic lung infections in patients suffering from cystic fibrosis as well as the major reason for morbidity in people with chronic obstructive pulmonary disease. P. aeruginosa infections are linked to diseases with high mortality rates and are challenging for treatment, for which no effective remedies have been developed. Massive lung epithelial cell death is a hallmark of severe acute lung injury and acute respiratory distress syndrome caused by P. aeruginosa infection. Lung epithelial cell death poses serious challenges to air barrier and structural integrity that may lead to edema, cytokine secretion, inflammatory infiltration, and hypoxia. Here we review different types of cell death caused by P. aeruginosa serving as a starting point for the diseases it is responsible for causing. We also review the different mechanisms of cell death and potential therapeutics in countering the serious challenges presented by this deadly bacterium.


Subject(s)
Acute Lung Injury , Epithelial Cells , Lung , Pseudomonas Infections , Pseudomonas aeruginosa , Respiratory Distress Syndrome , Acute Lung Injury/immunology , Acute Lung Injury/metabolism , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Cell Death/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/pathology , Humans , Lung/immunology , Lung/metabolism , Lung/microbiology , Lung/pathology , Pseudomonas Infections/immunology , Pseudomonas Infections/metabolism , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/metabolism , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/microbiology , Respiratory Distress Syndrome/pathology
20.
Front Immunol ; 11: 1722, 2020.
Article in English | MEDLINE | ID: mdl-32849610

ABSTRACT

The lung is a primary organ for gas exchange in mammals that represents the largest epithelial surface in direct contact with the external environment. It also serves as a crucial immune organ, which harbors both innate and adaptive immune cells to induce a potent immune response. Due to its direct contact with the outer environment, the lung serves as a primary target organ for many airborne pathogens, toxicants (aerosols), and allergens causing pneumonia, acute respiratory distress syndrome (ARDS), and acute lung injury or inflammation (ALI). The current review describes the immunological mechanisms responsible for bacterial pneumonia and sepsis-induced ALI. It highlights the immunological differences for the severity of bacterial sepsis-induced ALI as compared to the pneumonia-associated ALI. The immune-based differences between the Gram-positive and Gram-negative bacteria-induced pneumonia show different mechanisms to induce ALI. The role of pulmonary epithelial cells (PECs), alveolar macrophages (AMs), innate lymphoid cells (ILCs), and different pattern-recognition receptors (PRRs, including Toll-like receptors (TLRs) and inflammasome proteins) in neutrophil infiltration and ALI induction have been described during pneumonia and sepsis-induced ALI. Also, the resolution of inflammation is frequently observed during ALI associated with pneumonia, whereas sepsis-associated ALI lacks it. Hence, the review mainly describes the different immune mechanisms responsible for pneumonia and sepsis-induced ALI. The differences in immune response depending on the causal pathogen (Gram-positive or Gram-negative bacteria) associated pneumonia or sepsis-induced ALI should be taken in mind specific immune-based therapeutics.


Subject(s)
Acute Lung Injury/immunology , Immunity, Innate , Lung/immunology , Pneumonia, Bacterial/immunology , Sepsis/immunology , Acute Lung Injury/metabolism , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Animals , Host-Pathogen Interactions , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Lung/metabolism , Lung/microbiology , Lung/pathology , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , Sepsis/metabolism , Sepsis/microbiology , Sepsis/pathology , Signal Transduction , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...