Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
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
2.
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
3.
Biosci Rep ; 40(5)2020 05 29.
Article in English | MEDLINE | ID: mdl-32420586

ABSTRACT

The present study aims to reveal the molecular mechanism of peroxisome proliferator-activated receptor γ (PPARγ) on sepsis-induced acute lung injury (ALI). To do that, the rat injury model was established using cecal ligation and perforation (CLP) method, followed by different treatments, and the rats were divided into Sham group, CLP group, CLP + rosiglitazone (PPARγ agonist) group, CLP + GW9662 (PPARγ inhibitor) group, CLP + bpV (phosphatase and tensin homolog (PTEN) inhibitor) group, CLP + GW9662 + bpV group. Compared with Sham group, the mRNA and protein expression levels of PPARγ were down-regulated, the inflammation levels were elevated, and the apoptosis was increased in CLP group. After treatment with rosiglitazone, the protein expression level of PPARγ was significantly up-regulated, the phosphorylation level of PTEN/ß-catenin pathway was decreased, the PTEN/ß-catenin pathway was inhibited, the lung injury, inflammation and apoptosis were reduced. The opposite effect was observed after treatment with GW9662. Besides, bpV inhibited PTEN/ß-catenin pathway, and relieved the lung tissue injury. The overexpression of PPARγ reduced inflammatory response and inhibited apoptosis in sepsis-induced ALI. Furthermore, PPARγ relieved the sepsis-induced ALI by inhibiting the PTEN/ß-catenin pathway.


Subject(s)
Acute Lung Injury/prevention & control , Lung/drug effects , PPAR gamma/agonists , PTEN Phosphohydrolase/metabolism , Rosiglitazone/pharmacology , Sepsis/drug therapy , beta Catenin/metabolism , Acute Lung Injury/enzymology , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Anilides/pharmacology , Animals , Apoptosis/drug effects , Disease Models, Animal , Lung/enzymology , Lung/microbiology , Lung/pathology , Male , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , PPAR gamma/metabolism , Phosphorylation , Pneumonia/enzymology , Pneumonia/microbiology , Pneumonia/prevention & control , Pulmonary Edema/enzymology , Pulmonary Edema/microbiology , Pulmonary Edema/prevention & control , Rats, Sprague-Dawley , Sepsis/enzymology , Sepsis/microbiology , Signal Transduction
4.
Cell Microbiol ; 21(10): e13085, 2019 10.
Article in English | MEDLINE | ID: mdl-31290210

ABSTRACT

Staphylococcus aureus is frequently isolated from patients with community-acquired pneumonia and acute respiratory distress syndrome (ARDS). ARDS is associated with staphylococcal phosphatidylinositol-specific phospholipase C (PI-PLC); however, the role of PI-PLC in the pathogenesis and progression of ARDS remains unknown. Here, we showed that recombinant staphylococcal PI-PLC possesses enzyme activity that causes shedding of glycosylphosphatidylinositol-anchored CD55 and CD59 from human umbilical vein endothelial cell surfaces and triggers cell lysis via complement activity. Intranasal infection with PI-PLC-positive S. aureus resulted in greater neutrophil infiltration and increased pulmonary oedema compared with a plc-isogenic mutant. Although indistinguishable proinflammatory genes were induced, the wild-type strain activated higher levels of C5a in lung tissue accompanied by elevated albumin instillation and increased lactate dehydrogenase release in bronchoalveolar lavage fluid compared with the plc- mutant. Following treatment with cobra venom factor to deplete complement, the wild-type strain with PI-PLC showed a reduced ability to trigger pulmonary permeability and tissue damage. PI-PLC-positive S. aureus induced the formation of membrane attack complex, mainly on type II pneumocytes, and reduced the level of CD55/CD59, indicating the importance of complement regulation in pulmonary injury. In conclusion, S. aureus PI-PLC sensitised tissue to complement activation leading to more severe tissue damage, increased pulmonary oedema, and ARDS progression.


Subject(s)
Bacterial Proteins/metabolism , Complement System Proteins/metabolism , Phosphoinositide Phospholipase C/metabolism , Pulmonary Edema/immunology , Pulmonary Edema/microbiology , Respiratory Distress Syndrome/microbiology , Staphylococcal Infections/immunology , Staphylococcus aureus/enzymology , Alveolar Epithelial Cells/enzymology , Alveolar Epithelial Cells/immunology , Alveolar Epithelial Cells/microbiology , Animals , Bacterial Proteins/genetics , CD55 Antigens/immunology , CD59 Antigens/immunology , Cytokines/metabolism , Glycosylphosphatidylinositols/immunology , Glycosylphosphatidylinositols/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred BALB C , Phosphoinositide Phospholipase C/genetics , Pulmonary Edema/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/metabolism , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism
5.
Acta Pharmacol Sin ; 40(8): 1040-1048, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30644422

ABSTRACT

Staphylococcus aureus (S. aureus) infection leads to a severe inflammatory response and causes acute lung injury (ALI), eventually threatening human life. Therefore, it is of importance to find an agent to inhibit inflammation and reduce ALI. Here, we found that costunolide, a sesquiterpene lactone, displays anti-inflammatory effects and ameliorates heat-killed S. aureus (HKSA)-induced pneumonia. Costunolide treatment attenuated HKSA-induced murine ALI in which pulmonary neutrophil infiltration was inhibited, lung edema was decreased, and the production of pro-inflammatory cytokines was significantly reduced. In addition, costunolide dose-dependently inhibited the generation of IL-6, TNF-α, IL-1ß, and keratinocyte-derived cytokine (KC), as well as the expression of iNOS, in HKSA-induced macrophages. Furthermore, costunolide attenuated the phosphorylation of p38 MAPK and cAMP response element-binding protein (CREB). Collectively, our findings suggested that costunolide is a promising agent for alleviating bacterial-induced ALI via the inhibition of the MAPK signaling pathways.


Subject(s)
Acute Lung Injury/drug therapy , Anti-Inflammatory Agents/therapeutic use , Macrophage Activation/drug effects , Sesquiterpenes/therapeutic use , Acute Lung Injury/microbiology , Animals , Cytokines/metabolism , Lung/microbiology , Lung/pathology , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Pneumonia, Staphylococcal/drug therapy , Pneumonia, Staphylococcal/microbiology , Pulmonary Edema/drug therapy , Pulmonary Edema/microbiology , Staphylococcus aureus
6.
Thorax ; 74(1): 43-50, 2019 01.
Article in English | MEDLINE | ID: mdl-30076187

ABSTRACT

BACKGROUND: We previously reported that microvesicles (MVs) released by human mesenchymal stem cells (MSC) were as effective as the cells themselves in both Escherichia coli lipopolysaccharide and live bacteria-induced acute lung injury (ALI) mice models. However, it remained unclear whether the biological effect of MSC MV can be applied to human ALI. METHODS: In the current study, we tested the therapeutic effects of MSC MVs in a well-established ex vivo perfused human model of bacterial pneumonia. Using human donor lungs not used for transplantation, we instilled E. coli bacteria intrabronchially and, 1 hour later, administered MSC MVs into the perfusate as therapy. RESULTS: After 6 hours, instillation of E. coli bacteria caused influx of inflammatory cells, which resulted in significant inflammation, lung protein permeability and pulmonary oedema formation. Administration of MSC MV significantly increased alveolar fluid clearance and reduced protein permeability and numerically lowered the bacterial load in the injured alveolus. The beneficial effect on bacterial killing was more pronounced with pretreatment of MSCs with a Toll-like receptor 3 agonist, polyinosinic:polycytidylic acid (Poly (I:C)), prior to the isolation of MVs. Isolated human alveolar macrophages had increased antimicrobial activity with MSC MV treatment in vitro as well. Although oxygenation and lung compliance levels were similar between injury and treatment groups, administration of MSC MVs numerically decreased median pulmonary artery pressure at 6 hours. CONCLUSIONS: In summary, MSC MVs increased alveolar fluid clearance and reduced lung protein permeability, and pretreatment with Poly (I:C) enhanced the antimicrobial activity of MVs in an ex vivo perfused human lung with severe bacteria pneumonia.


Subject(s)
Acute Lung Injury/physiopathology , Acute Lung Injury/therapy , Cell- and Tissue-Based Therapy , Cell-Derived Microparticles , Escherichia coli Infections/complications , Mesenchymal Stem Cells , Pneumonia, Bacterial/complications , Proteins/metabolism , Pulmonary Alveoli/metabolism , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Adult , Aged , Arterial Pressure , Bacterial Load , Cell-Derived Microparticles/drug effects , Female , Humans , Interferon Inducers/pharmacology , Leukocyte Count , Lung Compliance , Male , Mesenchymal Stem Cells/drug effects , Middle Aged , Neutrophils , Organ Culture Techniques , Oxygen/metabolism , Permeability , Poly I-C/pharmacology , Pulmonary Alveoli/microbiology , Pulmonary Alveoli/pathology , Pulmonary Artery , Pulmonary Edema/microbiology , Pulmonary Edema/therapy , Toll-Like Receptor 3/agonists , Tumor Necrosis Factor-alpha/metabolism
7.
Toxicol Appl Pharmacol ; 364: 22-28, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30528763

ABSTRACT

Klebsiella pneumoniae causes severe infections including pneumonia and sepsis and treatments are complicated by increased levels of antibiotic resistance. We have identified a flavonoid kaempferol-3-O-glucorhamnoside derived from the plant Thesium chinense Turcz that possessed potent anti-inflammatory effects in K. pneumoniae infected mice. Administration of kaempferol-3-O-glucorhamnoside before bacterial challenge effectively suppressed expression of the major inflammatory cytokines TNF-α, IL-6, IL-1ß and PGE2 and ameliorated lung edema. In addition, administration of this compound to cultured RAW macrophages or Balb/c mice resulted in the suppression of NFκB and MAP kinase phosphorylation indicating an inhibitory effect on inflammation in vitro and in vivo. Kaempferol-3-O-glucorhamnoside also decreased ROS levels and overall oxidative stress in lungs and in cultured cells generated by K. pneumoniae exposure. Taken together, kaempferol-3-O-glucorhamnoside is a potent anti-inflammatory in vitro and in vivo and is a promising therapeutic agent for treating K. pneumoniae infections in the clinic.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Kaempferols/pharmacology , Klebsiella Infections/drug therapy , Klebsiella pneumoniae/pathogenicity , Lung/drug effects , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Pneumonia, Bacterial/drug therapy , Animals , Antioxidants/pharmacology , Dinoprostone/metabolism , Disease Models, Animal , Host-Pathogen Interactions , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Klebsiella Infections/enzymology , Klebsiella Infections/microbiology , Lung/enzymology , Lung/microbiology , Macrophages/drug effects , Macrophages/enzymology , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Pneumonia, Bacterial/enzymology , Pneumonia, Bacterial/microbiology , Pulmonary Edema/enzymology , Pulmonary Edema/microbiology , Pulmonary Edema/prevention & control , RAW 264.7 Cells , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
8.
Med Sci Monit ; 23: 1355-1362, 2017 Mar 18.
Article in English | MEDLINE | ID: mdl-28315572

ABSTRACT

BACKGROUND The aim of this study was to investigate the protective effects of cucurbitacin B (CuB) on sepsis-induced acute lung injury (ALI) in rats. MATERIAL AND METHODS An ALI model was made by cecal ligation and puncture (CLP) in SD rats. Rats were randomly divided into 5 groups (n=15 per group): animals undergoing a sham CLP (sham group); animals undergoing CLP (CLP control group); animals undergoing CLP and treated with CuB at 1 mg/kg of body weight (bw) (low-dose CuB [L-CuB] group), animals undergoing CuB at 2 mg/kg of bw (mid-dose CuB [M-CuB] group); and animals undergoing CuB at 5 mg/kg of bw (high-dose CuB [H-CuB] group). Samples of blood and lung tissue were harvested at different time points (6, 12, and 24 hour post-CLP surgery) for the detection of indicators which represented ALI. Five rats were respectively sacrificed at each time point. Pathological changes of lung tissue were observed by H&E staining. Another 50 rats were distributed into the same five groups to record the 72 hour survival rates. RESULTS Treatment with CuB significantly increased the blood gas PaO2 levels and decreased lung wet/dry (W/D) ratio (p<0.05). It significantly reduced protein concentration, accumulation of the inflammatory cells, and levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), (p<0.05), in the bronchoalveolar lavage fluid (BALF). Pulmonary pathological damage and survival rates at 72 hours were found to be effectively improved by CuB. In addition, CuB performed its pulmonary protection effects in a dose-depended manner. CONCLUSIONS CuB can effectively improve the pulmonary gas exchange function, reduce pulmonary edema, and inhibit the inflammatory response in the lung, revealing that CuB may serve as a potential therapeutic strategy for sepsis-induced ALI.


Subject(s)
Acute Lung Injury/drug therapy , Sepsis/drug therapy , Triterpenes/pharmacology , Acute Lung Injury/blood , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Animals , Bronchoalveolar Lavage Fluid , Cecum/injuries , Cecum/surgery , Disease Models, Animal , Interleukin-6/blood , Ligation , Lung/pathology , Male , Pulmonary Edema/blood , Pulmonary Edema/drug therapy , Pulmonary Edema/microbiology , Pulmonary Edema/pathology , Rats , Rats, Sprague-Dawley , Sepsis/pathology , Tumor Necrosis Factor-alpha/blood
9.
Arterioscler Thromb Vasc Biol ; 36(2): 339-49, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26663395

ABSTRACT

OBJECTIVE: Vascular endothelial (VE)-cadherin is the predominant component of endothelial adherens junctions essential for cell-cell adhesion and formation of the vascular barrier. Endocytic recycling is an important mechanism for maintaining the expression of cell surface membrane proteins. However, little is known about the molecular mechanism of VE-cadherin recycling and its role in maintenance of vascular integrity. APPROACH AND RESULTS: Using calcium-switch assay, confocal imaging, cell surface biotinylation, and flow cytometry, we showed that VE-cadherin recycling required Ras-related proteins in brain (Rab)11a and Rab11 family-interacting protein 2. Yeast 2-hybrid assay and coimmunoprecipitation demonstrated that direct interaction of VE-cadherin with family-interacting protein 2 (at aa 453-484) formed a ternary complex with Rab11a in human endothelial cells. Silencing of Rab11a or Rab11 family-interacting protein 2 in endothelial cells prevented VE-cadherin recycling and VE-cadherin expression at endothelial plasma membrane. Furthermore, inactivation of Rab11a signaling blocked junctional reannealing after vascular inflammation. Selective knockdown of Rab11a in pulmonary microvessels markedly increased vascular leakage in mice challenged with lipopolysaccharide or polymicrobial sepsis. CONCLUSIONS: Rab11a/Rab11 family-interacting protein 2-mediated VE-cadherin recycling is required for formation of adherens junctions and restoration of VE barrier integrity and hence a potential target for clinical intervention in inflammatory disease.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Capillary Permeability , Endocytosis , Endothelial Cells/enzymology , Lung/blood supply , Pulmonary Edema/metabolism , rab GTP-Binding Proteins/metabolism , Adherens Junctions/metabolism , Adherens Junctions/pathology , Animals , Antigens, CD/genetics , Cadherins/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Disease Models, Animal , Endothelial Cells/pathology , Endotoxemia/metabolism , Endotoxemia/microbiology , HEK293 Cells , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Protein Binding , Protein Stability , Protein Transport , Pulmonary Edema/microbiology , Pulmonary Edema/pathology , RNA Interference , Sepsis/metabolism , Sepsis/microbiology , Signal Transduction , Time Factors , Transfection , rab GTP-Binding Proteins/genetics
10.
Neuropathology ; 35(4): 343-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25955768

ABSTRACT

Neurogenic pulmonary edema (NPE) is a clinical syndrome characterized by the acute onset of pulmonary edema following a significant central nervous system insult. Only a few cases of NPE after Cryptococcal meningitis have been reported. We report a case of NPE following Cryptococcal meningoencephalitis. A 40-year-old man with no medical history was hospitalized for disturbance of consciousness. Blood glucose level was 124 mg/dL. Non-contrast head computed tomography showed no abnormalities. Lumbar puncture revealed a pressure of over 300 mm H2 O and cerebrospinal fluid (CSF) confirmed a white blood cell count of 65/mm(3) . The CSF glucose level was 0 mg/dL. The patient was empirically started on treatment for presumptive bacterial and viral meningitis. Four days after, the patient died in a sudden severe pulmonary edema. Autopsy was performed. We found at autopsy a brain edema with small hemorrhage of the right basal ganglia, severe pulmonary edema and mild cardiomegaly. Histologically, dilated Virchow-Robin spaces, crowded with Cryptococci were observed. In the right basal ganglia, Virchow-Robin spaces were destroyed with hemorrhage and Cryptococci spread to parenchyma of the brain. No inflammatory reaction of the lung was seen. Finally, acute pulmonary edema in this case was diagnosed as NPE following Cryptococcal meningoencephalitis. After autopsy, we found that he was positive for serum antibodies to human immunodeficiency virus.


Subject(s)
HIV Infections/complications , Meningitis, Cryptococcal/pathology , Meningoencephalitis/pathology , Pulmonary Edema/pathology , Adult , Antibodies , Cryptococcus neoformans/isolation & purification , Fatal Outcome , HIV Infections/blood , HIV Infections/immunology , Humans , Male , Meningoencephalitis/microbiology , Pulmonary Edema/microbiology
11.
J Mol Med (Berl) ; 93(6): 675-89, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25616357

ABSTRACT

UNLABELLED: Pulmonary edema associated with increased vascular permeability is a severe complication of Staphylococcus aureus-induced sepsis and an important cause of human pathology and death. We investigated the role of the mammalian acid sphingomyelinase (Asm)/ceramide system in the development of lung edema caused by S. aureus. Our findings demonstrate that genetic deficiency or pharmacologic inhibition of Asm reduced lung edema in mice infected with S. aureus. The Asm/ceramide system triggered the formation of superoxide, resulting in degradation of tight junction proteins followed by lung edema. Treatment of infected mice with amitriptyline, a potent inhibitor of Asm, protected mice from lung edema caused by S. aureus, but did not reduce systemic bacterial numbers. In turn, treatment with antibiotics reduced bacterial numbers but did not protect mice from lung edema. In contrast, only the combination of antibiotics and amitriptyline inhibited both pulmonary edema and bacteremia protecting mice from lethal sepsis and lung dysfunction suggesting the combination of both drugs as novel treatment option for sepsis. KEY MESSAGES: Antibiotics are often insufficient to cure S. aureus-induced sepsis. S. aureus induces lung edema via the Asm/ceramide system. Genetic deficiency of Asm inhibits lung dysfunction upon infection with S. aureus. Pharmacologic inhibition of Asm reduces lung edema induced by S. aureus. Antibiotics plus amitriptyline protect mice from lung edema and lethal S. aureus sepsis.


Subject(s)
Lung/drug effects , Pulmonary Edema/therapy , Sepsis/therapy , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Staphylococcal Infections/therapy , Staphylococcus aureus/drug effects , Amitriptyline/therapeutic use , Animals , Anti-Bacterial Agents/therapeutic use , Ceramides/metabolism , Enzyme Inhibitors/therapeutic use , Gene Knockout Techniques , Lung/metabolism , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Edema/genetics , Pulmonary Edema/microbiology , Pulmonary Edema/pathology , Sepsis/genetics , Sepsis/microbiology , Sepsis/pathology , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/metabolism , Staphylococcal Infections/complications , Staphylococcal Infections/genetics , Staphylococcal Infections/pathology , Superoxides/antagonists & inhibitors , Superoxides/metabolism
12.
Am J Respir Crit Care Med ; 190(5): 522-32, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25029038

ABSTRACT

RATIONALE: Alveolar liquid clearance is regulated by Na(+) uptake through the apically expressed epithelial sodium channel (ENaC) and basolaterally localized Na(+)-K(+)-ATPase in type II alveolar epithelial cells. Dysfunction of these Na(+) transporters during pulmonary inflammation can contribute to pulmonary edema. OBJECTIVES: In this study, we sought to determine the precise mechanism by which the TIP peptide, mimicking the lectin-like domain of tumor necrosis factor (TNF), stimulates Na(+) uptake in a homologous cell system in the presence or absence of the bacterial toxin pneumolysin (PLY). METHODS: We used a combined biochemical, electrophysiological, and molecular biological in vitro approach and assessed the physiological relevance of the lectin-like domain of TNF in alveolar liquid clearance in vivo by generating triple-mutant TNF knock-in mice that express a mutant TNF with deficient Na(+) uptake stimulatory activity. MEASUREMENTS AND MAIN RESULTS: TIP peptide directly activates ENaC, but not the Na(+)-K(+)-ATPase, upon binding to the carboxy-terminal domain of the α subunit of the channel. In the presence of PLY, a mediator of pneumococcal-induced pulmonary edema, this binding stabilizes the ENaC-PIP2-MARCKS complex, which is necessary for the open probability conformation of the channel and preserves ENaC-α protein expression, by means of blunting the protein kinase C-α pathway. Triple-mutant TNF knock-in mice are more prone than wild-type mice to develop edema with low-dose intratracheal PLY, correlating with reduced pulmonary ENaC-α subunit expression. CONCLUSIONS: These results demonstrate a novel TNF-mediated mechanism of direct ENaC activation and indicate a physiological role for the lectin-like domain of TNF in the resolution of alveolar edema during inflammation.


Subject(s)
Epithelial Sodium Channel Agonists/metabolism , Epithelial Sodium Channels/metabolism , Peptides, Cyclic/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Edema/metabolism , Streptolysins , Tumor Necrosis Factor-alpha/metabolism , Animals , Bacterial Proteins , Epithelial Sodium Channel Agonists/chemistry , Epithelial Sodium Channels/chemistry , Female , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Peptides, Cyclic/chemistry , Pulmonary Alveoli/microbiology , Pulmonary Edema/microbiology , Tumor Necrosis Factor-alpha/chemistry
13.
Expert Opin Drug Deliv ; 11(11): 1697-712, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25046368

ABSTRACT

OBJECTIVE: The objective of this study is to develop a nanostructured parenteral delivery system, laden with curcumin (CUR), for the therapeutic intervention of sepsis and associated pathologies. METHODS: Nanoemulsions were fabricated using sonication and speed homogenization. Size and zeta potential were evaluated by dynamic light scattering and transmission electron microscopy analysis. Pharmacodynamic and pharmacokinetic studies were performed on a rat model of lipopolysaccharide (LPS)-induced sepsis. RESULTS: The drug content of optimized nanoemulsion (F5) formulation (particle size 246 ± 08 nm, polydispersity index (PDI) of 0.120, zeta potential of -41.1 ± 1.2 mV) was found to be 1.25 mg/ml. In vitro release studies demonstrated that F5 was able to sustain the release of CUR for up to 24 h. Minimal hemolysis and cellular toxicity demonstrated its suitability for intravenous administration. Significant reduction of inflammatory mediator levels was mediated through enhanced uptake by in RAW 264.7 and THP-1 in absence/presence of LPS. Nanoemulsion resulted in an improvement of plasma concentration (AUCF5/AUC CUR = 8.80) and tissue distribution of CUR in rats leading to a reduction in LPS-induced lung and liver injury due to less neutrophil migration, reduced TNF-α levels and oxidative stress (demonstrated by levels of lipid peroxides as well as carbonylated proteins) as confirmed by histopathological studies. CONCLUSION: The findings suggest that the therapeutic performance (i.e., reduction in oxidative damage in tissues) of CUR can be enhanced by employing tocol acetate nanoemulsions (via improving pharmacokinetics and tissue distribution) as a platform for drug delivery in sepsis-induced organ injury.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Curcumin/administration & dosage , Disease Models, Animal , Drug Delivery Systems , Sepsis/drug therapy , Vitamins/chemistry , alpha-Tocopherol/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Cell Line , Curcumin/pharmacokinetics , Cytokines/blood , Drug Carriers , Emulsions/chemistry , Enzyme-Linked Immunosorbent Assay , Escherichia coli , Flow Cytometry , Infusions, Parenteral , Lipid Peroxides/metabolism , Lipopolysaccharides , Liver Diseases/blood , Liver Diseases/drug therapy , Liver Diseases/microbiology , Male , Nanoparticles/chemistry , Pulmonary Edema/blood , Pulmonary Edema/drug therapy , Pulmonary Edema/microbiology , Rats , Rats, Sprague-Dawley , Sepsis/blood , Sepsis/microbiology , Tissue Distribution
14.
Thorax ; 69(9): 819-25, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24891325

ABSTRACT

BACKGROUND: Human bone marrow-derived mesenchymal stem (stromal) cells (hMSCs) improve survival in mouse models of acute respiratory distress syndrome (ARDS) and reduce pulmonary oedema in a perfused human lung preparation injured with Escherichia coli bacteria. We hypothesised that clinical grade hMSCs would reduce the severity of acute lung injury (ALI) and would be safe in a sheep model of ARDS. METHODS: Adult sheep (30-40 kg) were surgically prepared. After 5 days of recovery, ALI was induced with cotton smoke insufflation, followed by instillation of live Pseudomonas aeruginosa (2.5×10(11) CFU) into both lungs under isoflurane anaesthesia. Following the injury, sheep were ventilated, resuscitated with lactated Ringer's solution and studied for 24 h. The sheep were randomly allocated to receive one of the following treatments intravenously over 1 h in one of the following groups: (1) control, PlasmaLyte A, n=8; (2) lower dose hMSCs, 5×10(6) hMSCs/kg, n=7; and (3) higher-dose hMSCs, 10×10(6) hMSCs/kg, n=4. RESULTS: By 24 h, the PaO2/FiO2 ratio was significantly improved in both hMSC treatment groups compared with the control group (control group: PaO2/FiO2 of 97±15 mm Hg; lower dose: 288±55 mm Hg (p=0.003); higher dose: 327±2 mm Hg (p=0.003)). The median lung water content was lower in the higher-dose hMSC-treated group compared with the control group (higher dose: 5.0 g wet/g dry [IQR 4.9-5.8] vs control: 6.7 g wet/g dry [IQR 6.4-7.5] (p=0.01)). The hMSCs had no adverse effects. CONCLUSIONS: Human MSCs were well tolerated and improved oxygenation and decreased pulmonary oedema in a sheep model of severe ARDS. TRAIL REGISTRATION NUMBER: NCT01775774 for Phase 1. NCT02097641 for Phase 2.


Subject(s)
Mesenchymal Stem Cell Transplantation , Pneumonia, Bacterial/complications , Pseudomonas Infections/complications , Pseudomonas aeruginosa , Pulmonary Edema/therapy , Respiratory Distress Syndrome/therapy , Administration, Intravenous , Animals , Aspartate Aminotransferases/blood , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Hemodynamics , Humans , Hypoxia/etiology , Hypoxia/physiopathology , Leukocyte Count , Neutrophils , Pulmonary Edema/microbiology , Pulmonary Edema/physiopathology , Random Allocation , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/physiopathology , Respiratory Function Tests , Severity of Illness Index , Sheep , Smoke Inhalation Injury/complications , Water-Electrolyte Balance
16.
World J Gastroenterol ; 20(16): 4771-7, 2014 Apr 28.
Article in English | MEDLINE | ID: mdl-24782631

ABSTRACT

AIM: To investigate whether mesenteric lymph from rats with severe intraperitoneal infection (SII) induces lung injury in healthy rats. METHODS: Twenty adult male specific pathogen-free Wistar rats were divided into two groups. Animals in the SII group received intraperitoneal injection of Escherichia coli (E. coli) at a dose of 0.3 mL/100 g. Control rats underwent the same procedure, but were injected with normal saline rather than E. coli. We ligated and drained the mesenteric lymphatic vessels and collected the mesenteric lymph. Mesenteric lymph collected from SII or control rats was infused intravenously into male healthy rats at a rate of 1 mL/h for 4 h. At the end of the infusion, all rats were sacrificed. Lungs were removed and examined histologically, and wet-to-dry weight (W/D) ratio and myeloperoxidase (MPO) activity were determined. Enzyme-linked immunosorbent assay (ELISA) was performed to determine the levels of the proinflammatory cytokines tumor necrosis factor (TNF)-α and interleukin (IL)-6. We performed Western blot to investigate the activation of Toll-like receptor (TLR)-4, and nuclear factor (NF)-κB p65. RESULTS: Compared with the control infusion group, there were obvious pathological changes in the SII group. The W/D ratio was significantly increased in the SII compared to control infusion group (5.86 ± 0.06 vs 5.37 ± 0.06, P < 0.01). MPO activity significantly increased in the SII infusion rats with a mean level of 0.86 ± 0.02 U/g compared to 0.18 ± 0.05 U/g in the control group (P < 0.01). The concentrations of TNF-α and IL-6 were significantly increased in the SII infusion group. The concentration of TNF-α was significantly increased in the SII infusion rats compared to control infusion rats (2104.46 ± 245.91 vs 1475.13 ± 137.82 pg/mL, P < 0.01). The concentration of IL-6 was significantly increased in the SII infusion rats with a mean level of 50.56 ± 2.85 pg/mL compared to 43.29 ± 2.02 pg/mL (P < 0.01). The expression levels of TLR-4 (7496.68 ± 376.43 vs 4589.02 ± 233.16, P < 0.01) and NF-κB (8722.19 ± 323.96 vs 6498.91 ± 338.76, P < 0.01) were significantly increased in the SII infusion group compared to the control infusion group. The infusion of SII lymph, but not control lymph, caused lung injury. CONCLUSION: The results indicate that SII lymph is sufficient to induce acute lung injury.


Subject(s)
Acute Lung Injury/etiology , Escherichia coli Infections/complications , Lung/metabolism , Lymph/metabolism , Peritonitis/complications , Sepsis/metabolism , Acute Lung Injury/metabolism , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Animals , Disease Models, Animal , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Inflammation Mediators/metabolism , Infusions, Intravenous , Interleukin-6/metabolism , Lung/microbiology , Lung/pathology , Lymph/microbiology , Male , Peritonitis/metabolism , Peritonitis/microbiology , Peroxidase/metabolism , Pulmonary Edema/metabolism , Pulmonary Edema/microbiology , Rats, Wistar , Sepsis/microbiology , Severity of Illness Index , Time Factors , Toll-Like Receptor 4/metabolism , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
17.
Toxins (Basel) ; 5(7): 1244-60, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23860351

ABSTRACT

Severe pneumonia is the main single cause of death worldwide in children under five years of age. The main etiological agent of pneumonia is the G+ bacterium Streptococcus pneumoniae, which accounts for up to 45% of all cases. Intriguingly, patients can still die days after commencing antibiotic treatment due to the development of permeability edema, although the pathogen was successfully cleared from their lungs. This condition is characterized by a dramatically impaired alveolar epithelial-capillary barrier function and a dysfunction of the sodium transporters required for edema reabsorption, including the apically expressed epithelial sodium channel (ENaC) and the basolaterally expressed sodium potassium pump (Na+-K+-ATPase). The main agent inducing this edema formation is the virulence factor pneumolysin, a cholesterol-binding pore-forming toxin, released in the alveolar compartment of the lungs when pneumococci are being lysed by antibiotic treatment or upon autolysis. Sub-lytic concentrations of pneumolysin can cause endothelial barrier dysfunction and can impair ENaC-mediated sodium uptake in type II alveolar epithelial cells. These events significantly contribute to the formation of permeability edema, for which currently no standard therapy is available. This review focuses on discussing some recent developments in the search for the novel therapeutic agents able to improve lung function despite the presence of pore-forming toxins. Such treatments could reduce the potentially lethal complications occurring after antibiotic treatment of patients with severe pneumonia.


Subject(s)
Lung/microbiology , Pneumonia/therapy , Streptococcus pneumoniae/pathogenicity , Streptolysins/toxicity , Animals , Bacterial Proteins/toxicity , Child, Preschool , Disease Models, Animal , Growth Hormone/metabolism , Humans , Immune System/microbiology , Lectins/therapeutic use , Lung/pathology , Pneumonia/microbiology , Pulmonary Edema/microbiology , Pulmonary Edema/therapy , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism , Virulence Factors
18.
Vet Pathol ; 50(6): 971-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23456966

ABSTRACT

Mycoplasma hyopneumoniae has a primary role in the porcine respiratory disease complex (PRDC). The objective of this study was to determine whether fumonisin mycotoxins influence the character and/or the severity of pathological processes induced in the lungs of pigs by Mycoplasma hyopneumoniae. Four groups of pigs (n = 7/group) were used, one fed 20 ppm fumonisin B1 (FB1) from 16 days of age (group F), one only infected with M. hyopneumoniae on study day 30 (group M), and a group fed FB1 and infected with M. hyopneumoniae (group MF), along with an untreated control group (group C). Computed tomography (CT) scans of infected pigs (M and MF) on study day 44 demonstrated lesions extending to the cranial and middle or in the cranial third of the caudal lobe of the lungs. The CT images obtained on study day 58 showed similar but milder lesions in 5 animals from group M, whereas lungs from 2 pigs in group MF appeared progressively worse. The evolution of average pulmonary density calculated from combined pixel frequency values, as measured by quantitative CT, was significantly influenced by the treatment and the age of the animals. The most characteristic histopathologic lesion in FB1-treated pigs was pulmonary edema, whereas the pathomorphological changes in Mycoplasma-infected pigs were consistent with catarrhal bronchointerstitial pneumonia. FB1 aggravated the progression of infection, as demonstrated by severe illness requiring euthanasia observed in 1 pig and evidence of progressive pathology in 2 pigs (group MF) between study days 44 and 58.


Subject(s)
Fumonisins/toxicity , Mycoplasma hyopneumoniae/isolation & purification , Pneumonia of Swine, Mycoplasmal/pathology , Pulmonary Edema/veterinary , Swine Diseases/pathology , Tomography, X-Ray Computed/veterinary , Animals , Disease Models, Animal , Lung/diagnostic imaging , Lung/drug effects , Lung/pathology , Mycotoxins/toxicity , Pneumonia of Swine, Mycoplasmal/diagnostic imaging , Pneumonia of Swine, Mycoplasmal/microbiology , Pulmonary Edema/microbiology , Pulmonary Edema/pathology , Random Allocation , Swine , Swine Diseases/chemically induced , Swine Diseases/microbiology
19.
Free Radic Biol Med ; 53(8): 1584-94, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22940620

ABSTRACT

Acute lung injury (ALI) initiates protective responses involving genes downstream of the Nrf2 (Nfe2l2) transcription factor, including heme oxygenase-1 (HO-1), which stimulates mitochondrial biogenesis and related anti-inflammatory processes. We examined mitochondrial biogenesis during Staphylococcus aureus pneumonia in mice and the effect of Nrf2 deficiency on lung mitochondrial biogenesis and resolution of lung inflammation. S. aureus pneumonia established by nasal insufflation of live bacteria was studied in mitochondrial reporter (mt-COX8-GFP) mice, wild-type (WT) mice, and Nrf2⁻/⁻ mice. Bronchoalveolar lavage, wet/dry ratios, real-time RT-PCR and Western analysis, immunohistochemistry, and fluorescence microscopy were performed on the lung at 0, 6, 24, and 48 h. The mice survived S. aureus inoculations at 5×108 CFU despite diffuse lung inflammation and edema, but the Nrf2⁻/⁻ lung showed increased ALI. In mt-COX8-GFP mice, mitochondrial fluorescence was enhanced in bronchial and alveolar type II (AT2) epithelial cells. WT mice displayed rapid HO-1 upregulation and lower proinflammatory TNF-α, IL-1ß, and CCL2 and, especially in AT2 cells, higher anti-inflammatory IL-10 and suppressor of cytokine signaling-3 than Nrf2⁻/⁻ mice. In the alveolar region, WT but not Nrf2⁻/⁻ mice showed strongly induced nuclear respiratory factor-1, PGC-1α, mitochondrial transcription factor-A, SOD2, Bnip3, mtDNA copy number, and citrate synthase. These findings indicate that S. aureus pneumonia induces Nrf2-dependent mitochondrial biogenesis in the alveolar region, mainly in AT2 cells. Absence of Nrf2 suppresses the alveolar transcriptional network for mitochondrial biogenesis and anti-inflammation, which worsens ALI. The findings link redox activation of mitochondrial biogenesis to ALI resolution.


Subject(s)
Acute Lung Injury/etiology , Mitochondrial Turnover , NF-E2-Related Factor 2/physiology , Pneumonia, Staphylococcal/complications , Pneumonia/etiology , Pulmonary Alveoli/pathology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Blotting, Western , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Female , Humans , Immunoenzyme Techniques , Inflammation Mediators/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidation-Reduction , Pneumonia/pathology , Pneumonia, Staphylococcal/microbiology , Pneumonia, Staphylococcal/pathology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/microbiology , Pulmonary Edema/metabolism , Pulmonary Edema/microbiology , Pulmonary Edema/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Staphylococcus aureus/pathogenicity , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
20.
Acta Cytol ; 56(1): 104-6, 2012.
Article in English | MEDLINE | ID: mdl-22236754

ABSTRACT

BACKGROUND: A well-known indication for the cytologic examination of bronchoalveolar lavage (BAL) fluid is the identification of infectious organisms. However, an important distinction must be made as to whether the organisms seen represent a true opportunistic lower respiratory tract infection or a non-pathologic contamination. CASE: We describe herein the case of a 13-month-old male infant who presented with persistent chest congestion and tracheobronchitis and who underwent BAL as part of his clinical work-up. On cytological examination of the BAL fluid, the Romanowsky-stained cytospin slides contained numerous squamous epithelial cells with some showing rare striated rod-like structures on their surfaces. The peculiar structures also had rounded ends and were very large when compared to adjacent known bacterial cocci. CONCLUSION: We have determined that the striated rod-like structures in the infant's BAL fluid were indeed bacteria, Simonsiella sp. Simonsiella has reportedly been found in up to 32% of oral swabs in normal humans and it is considered a commensal and non-pathogenic organism. The characteristically large size, the association with normal oral-derived squamous cells and the striated appearance is diagnostic and will hopefully eliminate any possibility of confusion with a truly pathogenic organism.


Subject(s)
Bacteroidetes/cytology , Bronchitis/microbiology , Bronchoalveolar Lavage Fluid/microbiology , Opportunistic Infections/microbiology , Pulmonary Edema/microbiology , Tracheitis/microbiology , Bacteroidetes/isolation & purification , Bronchitis/diagnosis , Diagnostic Errors , Humans , Infant , Male , Opportunistic Infections/complications , Opportunistic Infections/diagnosis , Pulmonary Edema/diagnosis , Tracheitis/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL
...