Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.000
Filter
Add more filters

Publication year range
1.
Toxicol Appl Pharmacol ; 480: 116742, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37923178

ABSTRACT

Acute hypobaric hypoxia at high altitude can cause fatal non-cardiogenic high altitude pulmonary edema. Anti-inflammatory and anti-oxidant treatments appear to be a prospective way to alleviate acute hypoxia lung injury. Kaempferol (KA) and ginsenoside Rg1 (GRg1) can be isolated and purified from ginseng with anti-inflammatory, antioxidant, anti-carcinogenic, neuroprotective, and antiaging effects. However, their effects and pharmacological mechanisms on lung injury remains unclear. Network pharmacology analyses were used to explore potential targets of KA and GRg1 against acute hypobaric hypoxia induced lung injury. Rat lung tissues were further used for animal experiment verification. Among the putative targets of KA and GRg1 for inhibition of acute hypobaric hypoxia induced lung injury, AKT1, PIK3R1, PTK2, STAT3, HSP90AA1 and AKT2 were recognized as higher interrelated targets. And PI3K-AKT signaling pathway is considered to be the most important and relevant pathway. The rat experimental results showed that KA and GRg1 significantly improved histopathological changes and decreased pulmonary edema in rats with lung injury caused by acute hypobaric hypoxia. The concentrations of IL-6, TNF-α, MDA, SOD and CAT in rats treated with KA and GRg1 were significantly ameliorated. Protein and mRNA levels of PI3K and AKTI were significantly inhibited after KA administration. KA and GRg1 can lower lung water content, improve lung tissue damage, reduce the production of pro-inflammatory cytokines and the oxidative stress level.


Subject(s)
Acute Lung Injury , Pulmonary Edema , Rats , Animals , Pulmonary Edema/drug therapy , Pulmonary Edema/prevention & control , Phosphatidylinositol 3-Kinases/metabolism , Kaempferols/pharmacology , Kaempferols/therapeutic use , Network Pharmacology , Hypoxia/complications , Hypoxia/drug therapy , Antioxidants , Acute Lung Injury/drug therapy , Anti-Inflammatory Agents
2.
Drug Chem Toxicol ; 46(5): 1024-1034, 2023 Nov.
Article in English | MEDLINE | ID: mdl-36069203

ABSTRACT

We aimed to observe the possible effects of melatonin (MLT) deprivation (pinealectomy) and exogenous MLT administration on pulmonary edema induced by alpha-naphthylthiourea (ANTU), a toxic chemical agent, in rats. Seventy animals were assigned to seven groups: control, sham pinealectomy (PINX), PINX, ANTU (10 mg/kg intraperitoneal on day 30), ANTU + MLT (10 mg/kg/day i.p. for 30 days), ANTU + PINX, and ANTU + PINX + MLT.In this study, pleural effusion (PE) formation, lung weight/body weight (LW/BW) and PE/BW ratios (fluid accumulation and weight values in the lungs) increase detected. Pre-ANTU MLT administration led to significant decreases in PE, LW/BW, and PE/BW levels. The inhibited glutathione (GSH) and superoxide dismutase (SOD) levels and high malondialdehyde (MDA) levels that ANTU increase lipid peroxidation in the study. MLT administration eliminated oxidative stress by reducing MDA and ameliorating GSH and SOD levels.Pre-ANTU MLT administration led to a significant decrease in interleukin-1 beta (IL-1ß) and tumor necrosis factor-alpha (TNF-α) levels in the lung when compared to the ANTU group without MLT administration. Post-pinealectomy ANTU administration significantly increased IL-1ß and TNF-α levels when compared to ANTU and MLT administration without pinealectomy. Diffused inflammatory cell infiltration, interstitial pulmonary edema, and histopathological congestion were observed after the administration of ANTU. Severity of the damage was elevated in the ANTU + PINX group. MLT treatment regressed pulmonary effusion and edema and improves lung structure. In brief, the findings suggested that MLT inhibited proinflammatory mediators and could serve as a therapeutic agent to prevent inflammatory disorders.


Subject(s)
Melatonin , Pulmonary Edema , Rats , Animals , Pulmonary Edema/chemically induced , Pulmonary Edema/prevention & control , Pulmonary Edema/pathology , Melatonin/pharmacology , Pinealectomy , Tumor Necrosis Factor-alpha , Thiourea/toxicity
3.
Pflugers Arch ; 474(10): 1053-1067, 2022 10.
Article in English | MEDLINE | ID: mdl-35778581

ABSTRACT

Acute hypoxia impairs left ventricular (LV) inotropic function and induces development of pulmonary edema (PE). Enhanced and uneven hypoxic pulmonary vasoconstriction is an important pathogenic factor of hypoxic PE. We hypothesized that the potent vasodilator relaxin might reduce hypoxic pulmonary vasoconstriction and prevent PE formation. Furthermore, as relaxin has shown beneficial effects in acute heart failure, we expected that relaxin might also improve LV inotropic function in hypoxia. Forty-two rats were exposed over 24 h to normoxia or hypoxia (10% N2 in O2). They were infused with either 0.9% NaCl solution (normoxic/hypoxic controls) or relaxin at two doses (15 and 75 µg kg-1 day-1). After 24 h, hemodynamic measurements and bronchoalveolar lavage were performed. Lung tissue was obtained for histological and immunohistochemical analyses. Hypoxic control rats presented significant depression of LV systolic pressure by 19% and of left and right ventricular contractility by about 40%. Relaxin did not prevent the hypoxic decrease in LV inotropic function, but re-increased right ventricular contractility. Moreover, hypoxia induced moderate interstitial PE and inflammation in the lung. Contrasting to our hypothesis, relaxin did not prevent hypoxia-induced pulmonary edema and inflammation. In hypoxic control rats, PE was similarly distributed in the apical and basal lung lobes. In relaxin-treated rats, PE index was 35-40% higher in the apical than in the basal lobe, which is probably due to gravity effects. We suggest that relaxin induced exaggerated vasodilation, and hence pulmonary overperfusion. In conclusion, the results show that relaxin does not prevent but rather may aggravate PE formation.


Subject(s)
Pulmonary Edema , Relaxin , Animals , Hypoxia/complications , Pneumonia/therapy , Pulmonary Artery , Pulmonary Edema/etiology , Pulmonary Edema/prevention & control , Rats , Relaxin/pharmacology , Relaxin/therapeutic use , Saline Solution/pharmacology , Vasodilator Agents/pharmacology
4.
Inflamm Res ; 71(2): 183-185, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34993559

ABSTRACT

Growth Hormone-Releasing Hormone (GHRH) is a neuropeptide regulating the release of Growth Hormone (GH) from the anterior pituitary gland, and acts as a growth factor in a diverse variety of tissues. GHRH antagonists (GHRHAnt) have been developed to counteract those events, and the beneficial effects of those peptides toward homeostasis have been associated with anti-inflammatory activities. Our lab is interested in delineating the mechanisms governing endothelial barrier function. Our goal is to establish new grounds on the development of efficient countermeasures against Acute Respiratory Distress Syndrome (ARDS), which has been associated with thousands of deaths worldwide due to COVID-19. Herein we demonstrate in vivo that GHRHAnt suppresses LPS-induced increase in bronchoalveolar lavage fluid (BALF) protein concentration, thus protecting the lungs against edema and inflammation.


Subject(s)
Bronchoalveolar Lavage Fluid/chemistry , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Lipopolysaccharides , Animals , COVID-19/complications , Growth Hormone-Releasing Hormone , Inflammation/etiology , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , Proteins/chemistry , Pulmonary Edema/etiology , Pulmonary Edema/prevention & control , Reactive Oxygen Species , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/etiology , SARS-CoV-2
5.
Am J Physiol Lung Cell Mol Physiol ; 320(4): L486-L497, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33439101

ABSTRACT

The constant transport of ions across the alveolar epithelial barrier regulates alveolar fluid homeostasis. Dysregulation or inhibition of Na+ transport causes fluid accumulation in the distal airspaces resulting in impaired gas exchange and respiratory failure. Previous studies have primarily focused on the critical role of amiloride-sensitive epithelial sodium channel (ENaC) in alveolar fluid clearance (AFC), yet activation of ENaC failed to attenuate pulmonary edema in clinical trials. Since 40% of AFC is amiloride-insensitive, Na+ channels/transporters other than ENaC such as Na+-coupled neutral amino acid transporters (SNATs) may provide novel therapeutic targets. Here, we identified a key role for SNAT2 (SLC38A2) in AFC and pulmonary edema resolution. In isolated perfused mouse and rat lungs, pharmacological inhibition of SNATs by HgCl2 and α-methylaminoisobutyric acid (MeAIB) impaired AFC. Quantitative RT-PCR identified SNAT2 as the highest expressed System A transporter in pulmonary epithelial cells. Pharmacological inhibition or siRNA-mediated knockdown of SNAT2 reduced transport of l-alanine across pulmonary epithelial cells. Homozygous Slc38a2-/- mice were subviable and died shortly after birth with severe cyanosis. Isolated lungs of Slc38a2+/- mice developed higher wet-to-dry weight ratios (W/D) as compared to wild type (WT) in response to hydrostatic stress. Similarly, W/D ratios were increased in Slc38a2+/- mice as compared to controls in an acid-induced lung injury model. Our results identify SNAT2 as a functional transporter for Na+ and neutral amino acids in pulmonary epithelial cells with a relevant role in AFC and the resolution of lung edema. Activation of SNAT2 may provide a new therapeutic strategy to counteract and/or reverse pulmonary edema.


Subject(s)
Acute Lung Injury/prevention & control , Amino Acid Transport System A/physiology , Extravascular Lung Water/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Edema/prevention & control , Sodium/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Hydrochloric Acid/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Rats , Rats, Sprague-Dawley
6.
Am J Physiol Lung Cell Mol Physiol ; 320(1): L17-L28, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33026237

ABSTRACT

Oxidative stress caused by mechanical ventilation contributes to the pathophysiology of ventilator-induced lung injury (VILI). A key mechanism maintaining redox balance is the upregulation of nuclear factor-erythroid-2-related factor 2 (Nrf2)-dependent antioxidant gene expression. We tested whether pretreatment with an Nrf2-antioxidant response element (ARE) pathway activator tert-butylhydroquinone (tBHQ) protects against VILI. Male C57BL/6J mice were pretreated with an intraperitoneal injection of tBHQ (n = 10), an equivalent volume of 3% ethanol (EtOH3%, vehicle, n = 13), or phosphate-buffered saline (controls, n = 10) and were then subjected to high tidal volume (HVT) ventilation for a maximum of 4 h. HVT ventilation severely impaired arterial oxygenation ([Formula: see text] = 49 ± 7 mmHg, means ± SD) and respiratory system compliance, resulting in a 100% mortality among controls. Compared with controls, tBHQ improved arterial oxygenation ([Formula: see text] = 90 ± 41 mmHg) and respiratory system compliance after HVT ventilation. In addition, tBHQ attenuated the HVT ventilation-induced development of lung edema and proinflammatory response, evidenced by lower concentrations of protein and proinflammatory cytokines (IL-1ß and TNF-α) in the bronchoalveolar lavage fluid, respectively. Moreover, tBHQ enhanced the pulmonary redox capacity, indicated by enhanced Nrf2-depentent gene expression at baseline and by the highest total glutathione concentration after HVT ventilation among all groups. Overall, tBHQ pretreatment resulted in 60% survival (P < 0.001 vs. controls). Interestingly, compared with controls, EtOH3% reduced the proinflammatory response to HVT ventilation in the lung, resulting in 38.5% survival (P = 0.0054 vs. controls). In this murine model of VILI, tBHQ increases the pulmonary redox capacity by activating the Nrf2-ARE pathway and protects against VILI. These findings support the efficacy of pharmacological Nrf2-ARE pathway activation to increase resilience against oxidative stress during injurious mechanical ventilation.


Subject(s)
Gene Expression Regulation , Hydroquinones/pharmacology , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Protective Agents/pharmacology , Pulmonary Edema/prevention & control , Ventilator-Induced Lung Injury/mortality , Animals , Antioxidant Response Elements , Antioxidants/pharmacology , Bronchoalveolar Lavage Fluid , Cytokines/metabolism , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , Pulmonary Edema/etiology , Respiration, Artificial/adverse effects , Survival Rate , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/etiology , Ventilator-Induced Lung Injury/pathology
7.
Toxicol Appl Pharmacol ; 422: 115460, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33774062

ABSTRACT

To explore the protective mechanism of simvastatin in acute lung injury (ALI), the lipopolysaccharide (LPS) induced (5 mg/kg) ALI rat model was used to examine the effects of simvastatin. Following simvastatin treatment, the histopathological evaluation of lung tissues was made using hematoxylin and eosin (H&E) staining. Also, myeloperoxidase (MPO) activity and the levels of tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1ß), and IL-10 were determined by ELISA. Blood gas analyses of arterial blood samples were performed to assess the pulmonary gas exchange. Moreover, the neutrophil count and total protein content were determined in the bronchoalveolar lavage (BAL) fluid. The ratio of wet lung to dry lung (W/D) and the alveolar fluid clearance (AFC) were calculated to estimate the severity of edema. Lastly, the levels of A2BAR, CFTR, claudin4, and claudin18 were also measured by qRT-PCR and Western blotting. Simvastatin treatment, in a dose-related manner, markedly improved the lung histological injury and decreased the levels of TNF-α, IL-1ß, and increased IL-10 in LPS induced ALI. Also, pulmonary neutrophil count was alleviated. Besides, a decreased ratio of W/D lung also confirmed the simvastatin intervention. Notably, simvastatin reduced the levels of A2BAR, CFTR, and claudin18 but upregulated claudin4 in lung tissues. Additionally, treatment with PSB1115, an antagonist of A2BAR, countered the protective effect of simvastatin in ALI. Our study demonstrates that simvastatin has a protective effect against LPS-induced ALI by activating A2BAR and should be exploited as a novel therapeutic target for the treatment of ALI.


Subject(s)
Acute Lung Injury/prevention & control , Adenosine A2 Receptor Agonists/pharmacology , Lung/drug effects , Receptor, Adenosine A2B/drug effects , Simvastatin/pharmacology , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Claudin-4/metabolism , Claudins/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cytokines/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Lipopolysaccharides , Lung/metabolism , Lung/pathology , Male , Neutrophil Infiltration/drug effects , Pulmonary Edema/chemically induced , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Pulmonary Edema/prevention & control , Rats, Sprague-Dawley , Receptor, Adenosine A2B/metabolism , Signal Transduction
8.
Am J Respir Crit Care Med ; 202(8): 1081-1087, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33054329

ABSTRACT

Protective ventilation strategies for the injured lung currently revolve around the use of low Vt, ostensibly to avoid volutrauma, together with positive end-expiratory pressure to increase the fraction of open lung and reduce atelectrauma. Protective ventilation is currently applied in a one-size-fits-all manner, and although this practical approach has reduced acute respiratory distress syndrome deaths, mortality is still high and improvements are at a standstill. Furthermore, how to minimize ventilator-induced lung injury (VILI) for any given lung remains controversial and poorly understood. Here we present a hypothesis of VILI pathogenesis that potentially serves as a basis upon which minimally injurious ventilation strategies might be developed. This hypothesis is based on evidence demonstrating that VILI begins in isolated lung regions manifesting a Permeability-Originated Obstruction Response (POOR) in which alveolar leak leads to surfactant dysfunction and increases local tissue stresses. VILI progresses topographically outward from these regions in a POOR-get-POORer fashion unless steps are taken to interrupt it. We propose that interrupting the POOR-get-POORer progression of lung injury relies on two principles: 1) open the lung to minimize the presence of heterogeneity-induced stress concentrators that are focused around the regions of atelectasis, and 2) ventilate in a patient-dependent manner that minimizes the number of lung units that close during each expiration so that they are not forced to rerecruit during the subsequent inspiration. These principles appear to be borne out in both patient and animal studies in which expiration is terminated before derecruitment of lung units has enough time to occur.


Subject(s)
Primary Prevention/methods , Pulmonary Atelectasis/prevention & control , Pulmonary Edema/prevention & control , Respiratory Distress Syndrome/physiopathology , Ventilator-Induced Lung Injury/prevention & control , Ventilator-Induced Lung Injury/physiopathology , Acute Disease , Biomechanical Phenomena , Chronic Disease , Female , Humans , Male , Monitoring, Physiologic , Prognosis , Pulmonary Atelectasis/etiology , Pulmonary Edema/etiology , Respiratory Distress Syndrome/therapy , Respiratory Function Tests
9.
Am J Physiol Lung Cell Mol Physiol ; 318(6): L1239-L1243, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32401673

ABSTRACT

Lethality of coronavirus disease (COVID-19) during the 2020 pandemic, currently still in the exponentially accelerating phase in most countries, is critically driven by disruption of the alveolo-capillary barrier of the lung, leading to lung edema as a direct consequence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We argue for inhibition of the transient receptor potential vanilloid 4 (TRPV4) calcium-permeable ion channel as a strategy to address this issue, based on the rationale that TRPV4 inhibition is protective in various preclinical models of lung edema and that TRPV4 hyperactivation potently damages the alveolo-capillary barrier, with lethal outcome. We believe that TRPV4 inhibition has a powerful prospect at protecting this vital barrier in COVID-19 patients, even to rescue a damaged barrier. A clinical trial using a selective TRPV4 inhibitor demonstrated a benign safety profile in healthy volunteers and in patients suffering from cardiogenic lung edema. We argue for expeditious clinical testing of this inhibitor in COVID-19 patients with respiratory malfunction and at risk for lung edema. Perplexingly, among the currently pursued therapeutic strategies against COVID-19, none is designed to directly protect the alveolo-capillary barrier. Successful protection of the alveolo-capillary barrier will not only reduce COVID-19 lethality but will also preempt a distressing healthcare scenario with insufficient capacity to provide ventilator-assisted respiration.


Subject(s)
Betacoronavirus , Coronavirus Infections , Lung/virology , Pandemics , Pneumonia, Viral , Pulmonary Edema/prevention & control , TRPV Cation Channels/antagonists & inhibitors , COVID-19 , Calcium/metabolism , Coronavirus Infections/virology , Humans , Lung/metabolism , Pneumonia, Viral/virology , Pulmonary Edema/virology , Respiration, Artificial , SARS-CoV-2
10.
Pharmacol Res ; 156: 104759, 2020 06.
Article in English | MEDLINE | ID: mdl-32200026

ABSTRACT

Acute lung injury (ALI), a severe and life-threatening inflammation of the lung, with high morbidity and mortality, underscoring the urgent need for novel treatments. Ge-Gen-Qin-Lian decoction (GQD), a classic Chinese herbal formula, has been widely used to treat intestine-related diseases in the clinic for centuries. In recent years, a growing number of studies have found that GQD has a favorable anti-inflammatory effect. With the further study on the viscera microbiota, the link between the lungs and the gut-the gut-lung axis has been established. Based on the theory of the gut-lung axis, we used systems pharmacology to explore the effects and mechanisms of GQD treatment in ALI. Hypothesizing that GQD inhibits ALI progression, we used the experimental model of lipopolysaccharide (LPS)-induced ALI in Balb/c mice to evaluate the therapeutic potential of GQD. Our results showed that GQD exerted protective effects against LPS-induced ALI by reducing pulmonary edema and microvascular permeability. Meanwhile, GQD can downregulate the expression of LPS-induced TNF-α, IL-1ß, and IL-6 in lung tissue, bronchoalveolar lavage fluid (BLAF), and serum. To further understand the molecular mechanism of GQD in the treatment of ALI, we used the network pharmacology to predict the disease targets of the active components of GQD. Lung tissue and serum samples of the mice were separately analyzed by transcriptomics and metabolomics. KEGG pathway analysis of network pharmacology and transcriptomics indicated that PI3K/Akt signaling pathway was significantly enriched, suggesting that it may be the main regulatory pathway for GQD treatment of ALI. By immunohistochemical analysis and apoptosis detection, it was verified that GQD can inhibit ALI apoptosis through PI3K/Akt signaling pathway. Then, we used the PI3K inhibitor LY294002 to block the PI3K/Akt signaling pathway, and reversely verified that the PI3K/Akt signaling pathway is the main pathway of GQD anti-ALI. In addition, differential metabolites in mice serum samples indicate that GQD can inhibit the inflammatory process of ALI by reversing the imbalance of energy metabolism. Our study showed that, GQD did have a better therapeutic effect on ALI, and initially elucidated its molecular mechanism. Thus, GQD could be exploited to develop novel therapeutics for ALI. Moreover, our study also provides a novel strategy to explore active components and effective mechanism of TCM formula combined with TCM theory to treat ALI.


Subject(s)
Acute Lung Injury/prevention & control , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Drugs, Chinese Herbal/pharmacology , Lung/drug effects , Systems Biology , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Regulatory Networks , Inflammation Mediators/metabolism , Lipopolysaccharides , Lung/metabolism , Lung/pathology , Male , Metabolomics , Mice, Inbred BALB C , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Protein Interaction Maps , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Edema/chemically induced , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Pulmonary Edema/prevention & control , Signal Transduction , Transcriptome
11.
Lung ; 198(2): 315-321, 2020 04.
Article in English | MEDLINE | ID: mdl-32086560

ABSTRACT

PURPOSE: Nicorandil is a hybrid between nitrates and KATP channel opener activators. The aim of this study was to evaluate the nicorandil's effects on ischemia-reperfusion (IR) lung injury and examine the mechanism of its effects. METHODS: Isolated rat lungs were divided into 6 groups. In the sham group, the lungs were perfused and ventilated for 150 min. In the IR group, after perfusion and ventilation for 30 min, they were interrupted (ischemia) for 60 min, and then resumed for 60 min. In the nicorandil (N) + IR group, nicorandil 6 mg was added before ischemia (nicorandil concentration was 75 µg ml-1). In the glibenclamide + N + IR group, the L-NAME (Nω-Nitro-L-arginine methyl ester) + N + IR group and ODQ (1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one) + N + IR group, glibenclamide 3 µM, L-NAME 100 µM, and ODQ 30 µM were added 5 min before nicorandil administration, respectively. We measured the coefficient of filtration (Kfc) of the lungs, total pulmonary vascular resistance, and the wet-to-dry lung weight ratio (WW/DW ratio). RESULTS: Kfc was significantly increased after 60 min reperfusion compared with baseline in the IR group, but no change in the sham group. An increase in Kfc was inhibited in the N + IR group compared with the IR group (0.92 ± 0.28 vs. 2.82 ± 0.68 ml min-1 mmHg-1 100 g-1; P < 0.01). Also, nicorandil attenuated WW/DW ratio was compared with IR group (8.3 ± 0.41 vs. 10.9 ± 2.5; P < 0.05). Nicorandil's inhibitory effect was blocked by glibenclamide and ODQ (P < 0.01), but not by L-NAME. CONCLUSIONS: Nicorandil attenuated IR injury in isolated rat lungs. This protective effect appears to involve its activation as KATP channel opener as well as that of the sGC-cGMP pathway.


Subject(s)
KATP Channels/agonists , Lung Injury/prevention & control , Lung/blood supply , Lung/drug effects , Membrane Transport Modulators/pharmacology , Nicorandil/pharmacology , Reperfusion Injury/prevention & control , Animals , Capillary Permeability/drug effects , Cyclic GMP/metabolism , KATP Channels/metabolism , Lung/metabolism , Lung/pathology , Lung Injury/metabolism , Lung Injury/pathology , Male , Perfusion , Pulmonary Circulation/drug effects , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Pulmonary Edema/prevention & control , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction , Soluble Guanylyl Cyclase/metabolism , Vascular Resistance/drug effects
12.
Heart Lung Circ ; 29(12): 1887-1892, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32327311

ABSTRACT

BACKGROUND: Pneumonectomy in the adult patient is associated with a mortality of 1-9%. Death is often due to post pneumonectomy pulmonary oedema (PPPO). The use of balanced chest drainage system (BCD) in the setting of post pneumonectomy has been reported to be of benefit in the prevention of PPPO. This study seeks to compare the incidence of PPPO in patients who underwent pneumonectomy and whose empty pleural space was managed either with CRD or BCD. METHODS: This retrospective observational cohort study involved 98 patients who were operated on by one surgeon at Liverpool Hospital, Sydney, Australia from 1997 to 2019. The patients were divided into two groups according to the era in which they had their pneumonectomy. Group 1 consisted of 18 patients managed with clamp-release drainage between 1997 and 2002. Group 2 consisted of 80 patients managed with balanced chest drainage between 2003 and 2019. The primary outcomes of interest were the development of PPPO and death. Demographic and clinico-pathological variables between the groups were compared including whether the phrenic nerve was sacrificed, volume of infused intraoperative fluid, duration of single lung ventilation, intraoperative tidal volumes, agents of anaesthetic induction and maintenance, mean urine output in the first 4 postoperative hours, institution of a postoperative 1.5 L fluid restriction, total chest drainage, day of chest drain removal, presence of radiological postoperative mediastinal shift, post-pneumonectomy pulmonary oedema and death. Group characteristics were compared using t-test and chi-squared for continuous and categorical variables respectively. Univariate and multivariate analysis was also undertaken using the Firth method of logistic regression for rare occurrences in a stepwise fashion. RESULTS: Through univariate analysis, balanced chest drainage, postoperative fluid restriction and intraoperative fluid infusion showed significant effect on PPPO. Through multivariate analysis, balanced chest drainage was found to have independent protective value for PPPO and mortality. CONCLUSION: Compared with clamp-release drainage, balanced chest drainage results in a lower incidence of post-pneumonectomy pulmonary oedema and death.


Subject(s)
Drainage/methods , Pneumonectomy/adverse effects , Postoperative Complications/prevention & control , Pulmonary Edema/prevention & control , Chest Tubes , Female , Follow-Up Studies , Humans , Male , Middle Aged , Postoperative Complications/etiology , Pulmonary Edema/etiology , Retrospective Studies , Thorax
13.
Undersea Hyperb Med ; 47(2): 189-196, 2020.
Article in English | MEDLINE | ID: mdl-32574434

ABSTRACT

Roflumilast is an inhibitor of phosphodiesterase-4 (PDE4) and can suppress the hydrolysis of cAMP in inflammatory cells, conferring anti-inflammatory effects. This study aimed to investigate the protective effects of roflumilast on hyperoxia-induced acute lung injury (HALI) in a rat model. Male Sprague-Dawley rats were randomly assigned into: control group; HALI group; 2.5 mg/kg roflumilast group; and 5 mg/kg roflumilast group. Rats were pressurized to 250 kPa with pure oxygen to induce lung injury. In the roflumilast groups, rats were orally administered with roflumilast at 2.5 or 5 mg/kg once before hyperoxia exposure and once daily for two days after exposure. Rats were sacrificed 72 hours after hyperoxia exposure. The lung tissues were collected for the detection of lung water content, inflammatory cytokines and NF-κB/p-NF-κB protein expression, and the bronchoalveolar lavage fluid was harvested for the measurement of protein concentration and lactate dehydrogenase activity. Results showed roflumilast at different doses could significantly reduce lung edema, improve lung pathology and reduce the expression of inflammatory cytokines in the lung. The protective effects seemed to be related to the dose of roflumilast. Our study indicates roflumilast has the potential as a medication for the treatment of HALI.


Subject(s)
Aminopyridines/therapeutic use , Benzamides/therapeutic use , Hyperoxia/complications , Lung Injury/prevention & control , Phosphodiesterase 4 Inhibitors/therapeutic use , Proteins/analysis , Aminopyridines/administration & dosage , Animals , Benzamides/administration & dosage , Body Water , Bronchoalveolar Lavage Fluid/chemistry , Cyclopropanes/administration & dosage , Cyclopropanes/therapeutic use , Interleukin-10/analysis , Interleukin-1beta/analysis , Interleukin-6/analysis , L-Lactate Dehydrogenase/analysis , Lung/chemistry , Lung/pathology , Lung Injury/etiology , Lung Injury/pathology , Male , NF-kappa B/analysis , Phosphodiesterase 4 Inhibitors/administration & dosage , Pulmonary Edema/prevention & control , Random Allocation , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/analysis
14.
J Cell Physiol ; 234(5): 6023-6032, 2019 05.
Article in English | MEDLINE | ID: mdl-30584668

ABSTRACT

Acute lung injury (ALI) is a life-threatening, diffuse heterogeneous lung injury characterized by acute onset, pulmonary edema, and respiratory failure. Lipopolysaccharide (LPS) is a leading cause for ALI and when administered to a mouse it induces a lung phenotype exhibiting some of the clinical characteristics of human ALI. This study focused on investigating whether microRNA-27b (miR-27b) affects ALI in a mouse model established by LPS-induction and to further explore the underlying mechanism. After model establishment, the mice were treated with miR-27b agomir, miR-27b antagomir, or D-ribofuranosylbenzimidazole (an inhibitor of nuclear factor-E2-related factor 2 [Nrf2]) to determine levels of miR-27b, Nrf2, nuclear factor kappa-light-chain-enhancer of activated B cells nuclear factor κB (NF-κB), p-NF-κB, and heme oxygenase-1 (HO-1). The levels of interleukin (IL)-1ß, IL-6, and tumor necrosis factor-α (TNF-α) in bronchoalveolar lavage fluid (BALF) were determined. The results of luciferase activity suggested that Nrf2 was a target gene of miR-27b. It was indicated that the Nrf2 level decreased in lung tissues from ALI mice. The downregulation of miR-27b decreased the levels of IL-1ß, IL-6, and TNF-α in BALF of ALI mice. Downregulated miR-27b increased Nrf2 level, thus enhancing HO-1 level along with reduction of NF-κB level as well as the extent of NF-κB phosphorylation in the lung tissues of the transfected mice. Pathological changes were ameliorated in LPS-reduced mice elicited by miR-27b inhibition. The results of this study demonstrate that downregulated miR-27b couldenhance Nrf2 and HO-1 expressions, inhibit NF-κB signaling pathway, which exerts a protective effect on LPS-induced ALI in mice.


Subject(s)
Acute Lung Injury/prevention & control , Antagomirs/pharmacology , Anti-Inflammatory Agents/pharmacology , Dichlororibofuranosylbenzimidazole/pharmacology , Lung/drug effects , MicroRNAs/antagonists & inhibitors , NF-E2-Related Factor 2/antagonists & inhibitors , NF-kappa B/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Cytokines/metabolism , Disease Models, Animal , Down-Regulation , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Lipopolysaccharides , Lung/metabolism , Lung/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , NF-kappa B/genetics , Pulmonary Edema/chemically induced , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Pulmonary Edema/prevention & control , Signal Transduction
15.
J Cell Physiol ; 234(5): 6012-6022, 2019 05.
Article in English | MEDLINE | ID: mdl-30589073

ABSTRACT

Spinal cord injury (SCI) is one kind of severe traumatic injury, resulting in systemic inflammatory response syndrome and secondary lung injury, which is an important pathological basis of respiratory complications. The nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome is an important cytosolic protein complex in many inflammatory diseases. Hence, it is inescapable to explore the effect of inhibition of NLRP3 inflammasome by inhibitors in a mouse SCI model, which was conducted by using the method of 30-G closing force aneurysm clipping at T6-T7 spinal segment for 1 min, followed by assessment of edema, histology, alveolar type II cell apoptosis, mitochondrial dysfunction, and neutrophil infiltration. In brief, our results showed that, NLRP3 inflammasome inhibitor BAY 11-7082 or A438079 inhibited activation of NLRP3 inflammasome, alleviated mitochondrial dysfunction, the number of macrophage and neutrophil, thereby attenuating alveolar type II cell apoptosis, lung edema, and histological injury. Taken together, our data reveal that NLRP3 inflammasome inhibitor BAY 11-7082 or A438079 attenuates the inflammatory response, reverses mitochondrial dysfunction, and subsequently alleviates secondary lung injury following SCI.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammasomes/antagonists & inhibitors , Lung Injury/prevention & control , Lung/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Nitriles/pharmacology , Pyridines/pharmacology , Spinal Cord Injuries/drug therapy , Sulfones/pharmacology , Tetrazoles/pharmacology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Apoptosis/drug effects , Disease Models, Animal , Female , Inflammasomes/immunology , Inflammasomes/metabolism , Lung/immunology , Lung/metabolism , Lung/pathology , Lung Injury/immunology , Lung Injury/metabolism , Lung Injury/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neutrophil Infiltration/drug effects , Pulmonary Edema/immunology , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Pulmonary Edema/prevention & control , Signal Transduction , Spinal Cord Injuries/immunology , Spinal Cord Injuries/metabolism , Time Factors
16.
Am J Physiol Renal Physiol ; 317(2): F343-F360, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31091126

ABSTRACT

Polycystic kidney disease (PKD) is characterized by slowly expanding renal cysts that damage the kidney, typically resulting in renal failure by the fifth decade. The most common cause of death in these patients, however, is cardiovascular disease. Expanding cysts in PKD induce chronic kidney injury that is accompanied by immune cell infiltration, including macrophages, which we and others have shown can promote disease progression in PKD mouse models. Here, we show that monocyte chemoattractant protein-1 [MCP-1/chemokine (C-C motif) ligand 2 (CCL2)] is responsible for the majority of monocyte chemoattractant activity produced by renal PKD cells from both mice and humans. To test whether the absence of MCP-1 lowers renal macrophage concentration and slows disease progression, we generated genetic knockout (KO) of MCP-1 in a mouse model of PKD [congenital polycystic kidney (cpk) mice]. Cpk mice are born with rapidly expanding renal cysts, accompanied by a decline in kidney function and death by postnatal day 21. Here, we report that KO of MCP-1 in these mice increased survival, with some mice living past 3 mo. Surprisingly, however, there was no significant difference in renal macrophage concentration, nor was there improvement in cystic disease or kidney function. Examination of mice revealed cardiac hypertrophy in cpk mice, and measurement of cardiac electrical activity via ECG revealed repolarization abnormalities. MCP-1 KO did not affect the number of cardiac macrophages, nor did it alleviate the cardiac aberrancies. However, MCP-1 KO did prevent the development of pulmonary edema, which occurred in cpk mice, and promoted decreased resting heart rate and increased heart rate variability in both cpk and noncystic mice. These data suggest that in this mouse model of PKD, MCP-1 altered cardiac/pulmonary function and promoted death outside of its role as a macrophage chemoattractant.


Subject(s)
Arrhythmias, Cardiac/metabolism , Cardiomegaly/metabolism , Chemokine CCL2/metabolism , Kidney/metabolism , Lung/metabolism , Myocardium/metabolism , Polycystic Kidney Diseases/metabolism , Pulmonary Edema/metabolism , Animals , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cells, Cultured , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Disease Models, Animal , Disease Progression , Fibrosis , Humans , Inflammation Mediators/metabolism , Kidney/pathology , Kidney/physiopathology , Lung/pathology , Lung/physiopathology , Macrophages/metabolism , Macrophages/pathology , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Polycystic Kidney Diseases/pathology , Polycystic Kidney Diseases/physiopathology , Pulmonary Edema/pathology , Pulmonary Edema/physiopathology , Pulmonary Edema/prevention & control , Time Factors
17.
Am J Transplant ; 19(2): 345-355, 2019 02.
Article in English | MEDLINE | ID: mdl-30106236

ABSTRACT

Ex vivo lung perfusion (EVLP) has become routine practice in lung transplantation. Still, running periods exceeding 12 hours have not been undertaken clinically to date, and it remains unclear how the perfusion solution for extended running periods should be composed and which parameters may predict outcomes. Twenty-four porcine lungs underwent EVLP for 24 hours using the Organ Care System (OCS). Lungs were ventilated and perfused with STEEN's solution enriched with erythrocytes (n = 8), acellular STEEN's solution (n = 8), or low-potassium dextran (LPD) solution enriched with erythrocytes (n = 8). After 24 hours, the left lungs were transplanted into recipient pigs. After clamping of the contralateral lung, the recipients were observed for 6 hours. The most favorable outcome was observed in organs utilizing STEEN solution enriched with erythrocytes as perfusate, whereas the least favorable outcome was seen with LPD solution enriched with erythrocytes for perfusion. Animals surviving the observation period showed lower peak airway pressure (PAWP) and pulmonary vascular resistance (PVR) during OCS preservation. The results suggest that transplantation of lungs following 24 hours of EVLP is feasible but dependent on the composition of the perfusate. PAWP and PVR during EVLP are early and late predictors of transplant outcome, respectively.


Subject(s)
Disease Models, Animal , Extracorporeal Circulation/methods , Lung Transplantation/methods , Lung/physiology , Organ Preservation/methods , Perfusion/methods , Pulmonary Edema/prevention & control , Animals , Organ Preservation Solutions/administration & dosage , Swine , Tissue Donors
18.
Microvasc Res ; 125: 103885, 2019 09.
Article in English | MEDLINE | ID: mdl-31175855

ABSTRACT

BACKGROUND: Successful amelioration of long-term warm ischemia lung injury in donors after cardiac death (DCDs) can remarkably improve outcomes. Hydrogen gas provides potent anti-inflammatory and antioxidant effects against ischemia-reperfusion injury (IRI). This study observed the effects of hydrogen inhalation on lung grafts during the warm ischemia phase in cardiac death donors. METHODS: After cardiac death, rat donor lungs (n = 8) underwent mechanical ventilation with 40% oxygen plus 60% nitrogen (control group) or 3% hydrogen and 40% oxygen plus 57% nitrogen (hydrogen group) for 2 h during the warm ischemia phase in situ. Then, lung transplantation was performed after 2 h of cold storage and 3 h of recipient reperfusion prior to lung graft assessment. Rats that underwent left thoracotomy without transplantation served as the sham group (n = 8). The results of static compliance and arterial blood gas analysis were assessed in the recipients. The wet-to-dry weight ratio (W/D), inflammation, oxidative stress, cell apoptosis and histologic changes were evaluated after 3 h of reperfusion. Nuclear factor kappa B (NF-κB) protein expression in the graft was analyzed by Western blotting. RESULTS: Compared with the sham group, lung function, W/D, inflammatory reaction, oxidative stress and histological changes were decreased in both transplant groups (control and hydrogen groups). However, compared with the control group, exposure to 3% hydrogen significantly improved lung graft static compliance and oxygenation and remarkably decreased the wet-to-dry weight ratio, inflammatory reactions, and lipid peroxidation. Furthermore, hydrogen improved the lung graft histological changes, decreased the lung injury score and apoptotic index and reduced NF-κB nuclear accumulation in the lung grafts. CONCLUSION: Lung inhalation with 3% hydrogen during the warm ischemia phase attenuated lung graft IRI via NF-κB-dependent anti-inflammatory and antioxidative effects in rat donors after cardiac death.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Hydrogen/administration & dosage , Lung Injury/prevention & control , Lung Transplantation , Lung/drug effects , Lung/surgery , Reperfusion Injury/prevention & control , Warm Ischemia , Administration, Inhalation , Animals , Apoptosis/drug effects , Disease Models, Animal , Inflammation Mediators/metabolism , Lipid Peroxidation/drug effects , Lung/metabolism , Lung/pathology , Lung Injury/etiology , Lung Injury/metabolism , Lung Injury/pathology , Lung Transplantation/adverse effects , Male , Oxidative Stress/drug effects , Pulmonary Edema/etiology , Pulmonary Edema/metabolism , Pulmonary Edema/pathology , Pulmonary Edema/prevention & control , Rats, Sprague-Dawley , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Time Factors , Warm Ischemia/adverse effects
19.
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
20.
Respir Res ; 20(1): 245, 2019 Nov 06.
Article in English | MEDLINE | ID: mdl-31694668

ABSTRACT

AIM: In acute respiratory distress syndrome (ARDS) damaged alveolar epithelium, leakage of plasma proteins into the alveolar space and inactivation of pulmonary surfactant lead to respiratory dysfunction. Lung function could potentially be restored with exogenous surfactant therapy, but clinical trials have so far been disappointing. These negative results may be explained by inactivation and/or too low doses of the administered surfactant. Surfactant based on a recombinant surfactant protein C analogue (rSP-C33Leu) is easy to produce and in this study we compared its effects on lung function and inflammation with a commercial surfactant preparation in an adult rabbit model of ARDS. METHODS: ARDS was induced in adult New Zealand rabbits by mild lung-lavages followed by injurious ventilation (VT 20 m/kg body weight) until P/F ratio < 26.7 kPa. The animals were treated with two intratracheal boluses of 2.5 mL/kg of 2% rSP-C33Leu in DPPC/egg PC/POPG, 50:40:10 or poractant alfa (Curosurf®), both surfactants containing 80 mg phospholipids/mL, or air as control. The animals were subsequently ventilated (VT 8-9 m/kg body weight) for an additional 3 h and lung function parameters were recorded. Histological appearance of the lungs, degree of lung oedema and levels of the cytokines TNFα IL-6 and IL-8 in lung homogenates were evaluated. RESULTS: Both surfactant preparations improved lung function vs. the control group and also reduced inflammation scores, production of pro-inflammatory cytokines, and formation of lung oedema to similar degrees. Poractant alfa improved compliance at 1 h, P/F ratio and PaO2 at 1.5 h compared to rSP-C33Leu surfactant. CONCLUSION: This study indicates that treatment of experimental ARDS with synthetic lung surfactant based on rSP-C33Leu improves lung function and attenuates inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Biological Products/pharmacology , Lung/drug effects , Phospholipids/pharmacology , Pneumonia/prevention & control , Pulmonary Surfactant-Associated Protein C/pharmacology , Pulmonary Surfactants/pharmacology , Respiratory Distress Syndrome/drug therapy , Animals , Cytokines/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Lung/metabolism , Lung/physiopathology , Pneumonia/metabolism , Pneumonia/physiopathology , Pulmonary Edema/metabolism , Pulmonary Edema/physiopathology , Pulmonary Edema/prevention & control , Rabbits , Recombinant Proteins/pharmacology , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL