Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
Cell Death Dis ; 14(6): 352, 2023 06 08.
Article in English | MEDLINE | ID: mdl-37291088

ABSTRACT

Idiopathic pulmonary fibrosis is a progressive fibrotic disorder with no cure that is characterized by deterioration of lung function. Current FDA-approved drugs for IPF delay the decline in lung function, but neither reverse fibrosis nor significantly improve overall survival. SHP-1 deficiency results in hyperactive alveolar macrophages accumulating in the lung, which contribute to the induction of pulmonary fibrosis. Herein, we investigated whether employing a SHP-1 agonist ameliorates pulmonary fibrosis in a bleomycin-induced pulmonary fibrosis murine model. Histological examination and micro-computed tomography images showed that SHP-1 agonist treatment alleviates bleomycin-induced pulmonary fibrosis. Reduced alveolar hemorrhage, lung inflammation, and collagen deposition, as well as enhanced alveolar space, lung capacity, and improved overall survival were observed in mice administered the SHP-1 agonist. The percentage of macrophages collected from bronchoalveolar lavage fluid and circulating monocytes in bleomycin-instilled mice were also significantly reduced by SHP-1 agonist treatment, suggesting that the SHP-1 agonist may alleviate pulmonary fibrosis by targeting macrophages and reshaping the immunofibrotic niche. In human monocyte-derived macrophages, SHP-1 agonist treatment downregulated CSF1R expression and inactivated STAT3/NFκB signaling, culminating in inhibited macrophage survival and perturbed macrophage polarization. The expression of pro-fibrotic markers (e.g., MRC1, CD200R1, and FN1) by IL4/IL13-induced M2 macrophages that rely on CSF1R signaling for their fate-determination was restricted by SHP-1 agonist treatment. While M2-derived medium promoted the expression of fibroblast-to-myofibroblast transition markers (e.g., ACTA2 and COL3A1), the application of SHP-1 agonist reversed the transition in a dose-dependent manner. Our report indicates that pharmacological activation of SHP-1 ameliorates pulmonary fibrosis via suppression of CSF1R signaling in macrophages, reduction of pathogenic macrophages, and the inhibition of fibroblast-to-myofibroblast transition. Our study thus identifies SHP-1 as a druggable target for the treatment of IPF, and suggests that the SHP-1 agonist may be developed as an anti-pulmonary fibrosis medication that both suppresses inflammation and restrains fibroblast-to-myofibroblast transition.


Subject(s)
Idiopathic Pulmonary Fibrosis , Macrophages , Mice , Humans , Animals , X-Ray Microtomography , Macrophages/metabolism , Lung/metabolism , Inflammation/pathology , Idiopathic Pulmonary Fibrosis/pathology , Bleomycin/therapeutic use , Fibrosis , Mice, Inbred C57BL
3.
Front Pharmacol ; 13: 1074986, 2022.
Article in English | MEDLINE | ID: mdl-36582541

ABSTRACT

Dysregulation of macrophages in the pro-inflammatory (M1) and anti-inflammatory (M2) sub-phenotypes is a crucial element in several inflammation-related diseases and injuries. We investigated the role of aquaporin (AQP) in macrophage polarization using AQP pan-inhibitor mercury chloride (HgCl2). Lipopolysaccharides (LPSs) induced the expression of AQP-1 and AQP-9 which increased the cell size of bone marrow-derived macrophages. The inhibition of AQPs by HgCl2 abolished cell size changes and significantly suppressed M1 polarization. HgCl2 significantly reduced the activation of the nuclear factor kappa B (NF-κB) and p38 mitogen-activated protein kinase (MAPK) pathways and inhibited the production of IL-1ß. HgCl2 attenuated LPS-induced activation of mitochondria and reactive oxygen species production and autophagy was promoted by HgCl2. The increase in the light chain three II/light chain three I ratio and the reduction in PTEN-induced kinase one expression suggests the recycling of damaged mitochondria and the restoration of mitochondrial activity by HgCl2. In summary, the present study demonstrates a possible mechanism of the AQP inhibitor HgCl2 in macrophage M1 polarization through the restriction of cell volume change, suppression of the p38 MAPK/NFκB pathway, and promotion of autophagy.

4.
Front Immunol ; 13: 916102, 2022.
Article in English | MEDLINE | ID: mdl-35812413

ABSTRACT

Background: Lipopolysaccharide (LPS)-induced acute lung injury (ALI) induces endoplasmic reticulum stress, unfolded protein response (UPR), apoptosis, and inflammation. Inositol-requiring enzyme 1 (IRE1)-α is important for adaptive and apoptotic UPR determination during ER stress. The aqueous extract of Descuraniae Semen (AEDS) is reported to be a safe and effective herb for the treatment of pulmonary edema as it shows anti-inflammatory activities. Methods: We investigated the effects of AEDS on LPS-induced ALI in A549 cells with respect to the regulation of IRE1α-dependent UPR, proteasomal degradation, mitochondrial membrane potential (MtMP), inflammation, and apoptosis. Results: AEDS attenuated ER stress by regulating the proteasomal degradation. LPS induced ER stress [binding immunoglobulin protein (BiP), phosphorylated IRE1α, sliced X-box binding protein 1 [XBP1s], phosphorylated cJUN NH2-terminal kinase (pJNK), B-cell lymphoma (Bcl)-2-associated X (Bax), Bcl-2], inflammation (nucleus factor-kappa B (NF-κB) p65 nuclear translocation, nucleus NF-κB, pro-inflammatory cytokines] and apoptosis [C/EBP homologous protein (CHOP), cytochrome c, caspase-8, and caspase-6, and TUNEL] were significantly attenuated by AEDS treatment in A549 cells. AEDS prevents LPS-induced decreased expression of MtMP in A549 cells. Conclusions: AEDS attenuated LPS-induced inflammation and apoptosis by regulating proteasomal degradation, promoting IRE1α-dependent adaptive UPR, and inhibiting IRE1α-dependent apoptotic UPR. Moreover, IRE1α-dependent UPR plays a pivotal role in the mechanisms of LPS-induced ALI. Based on these findings, AEDS is suggested as a potential therapeutic option for treating patients with ALI.


Subject(s)
Acute Lung Injury , Lipopolysaccharides , A549 Cells , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Apoptosis , Endoribonucleases/metabolism , Humans , Inflammation/chemically induced , Inflammation/drug therapy , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Protein Serine-Threonine Kinases , Semen/metabolism , Unfolded Protein Response
5.
Biomedicines ; 10(4)2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35453586

ABSTRACT

BACKGROUND: In the management of major burn wounds, allogeneic skin transplantation is a critical procedure to improve wound repair. Our previous works found that intermittent exposure to carbon dioxide leads to permissive hypercapnia (HCA) and prolongs skin allograft survival. However, the modulatory effects of HCA exposure on the immune system are not well understood. OBJECTIVES: Our purpose was to investigate how intermittent exposure to HCA can effectively reduce the immune reaction to allogeneic skin graft rejection. METHODS: A fully major histocompatibility complex-incompatible skin transplant from BALB/c to C57BL/6 mice model was utilized. Immune cells from splenic and draining lymph nodes were analyzed by flow cytometry. Serum proinflammatory cytokines were analyzed by ELISA. RESULTS: Serum levels of IFN-γ, IL-2, IL-6, and TNF-α were significantly decreased in the HCA group. Additionally, the percentage of CD8+ cells in draining lymph nodes was significantly lower in HCA than in the control group. Moreover, the generation rate of FoxP3+ regulatory T cells (Tregs) from spleen naïve CD4+ T cells was increased by intermittent exposure to carbon dioxide. The infiltrated neutrophils were also eliminated by HCA. Taken together, we concluded that intermittent hypercapnia exposure could effectively suppress skin rejection by stimulating Treg cell generation and suppressing immune reactions.

6.
Front Immunol ; 12: 720844, 2021.
Article in English | MEDLINE | ID: mdl-34489970

ABSTRACT

Background: Ventilator-induced lung injury (VILI) is characterized by vascular barrier dysfunction and suppression of alveolar fluid clearance (AFC). Obesity itself leads to chronic inflammation, which may initiate an injurious cascade to the lungs and simultaneously induce a protective feedback. In this study, we investigated the protective mechanism of obesity on VILI in a mouse model. Methods: The VILI model was set up via 6-h mechanical ventilation with a high tidal volume. Parameters including lung injury score, STAT3/NFκB pathway, and AFC were assessed. Mice with diet-induced obesity were obtained by allowing free access to a high-fat diet since the age of 3 weeks. After a 9-week diet intervention, these mice were sacrificed at the age of 12 weeks. The manipulation of SOCS3 protein was achieved by siRNA knockdown and pharmaceutical stimulation using hesperetin. WNK4 knockin and knockout obese mice were used to clarify the pathway of AFC modulation. Results: Obesity itself attenuated VILI. Knockdown of SOCS3 in obese mice offset the protection against VILI afforded by obesity. Hesperetin stimulated SOCS3 upregulation in nonobese mice and provided protection against VILI. In obese mice, the WNK4 axis was upregulated at the baseline, but was significantly attenuated after VILI compared with nonobese mice. At the baseline, the manipulation of SOCS3 by siRNA and hesperetin also led to the corresponding alteration of WNK4, albeit to a lesser extent. After VILI, WNK4 expression correlated with STAT3/NFκB activation, regardless of SOCS3 status. Obese mice carrying WNK4 knockout had VILI with a severity similar to that of wild-type obese mice. The severity of VILI in WNK4-knockin obese mice was counteracted by obesity, similar to that of wild-type nonobese mice only. Conclusions: Obesity protects lungs from VILI by upregulating SOCS3, thus suppressing the STAT3/NFκB inflammatory pathway and enhancing WNK4-related AFC. However, WNK4 activation is mainly from direct NFκB downstreaming, and less from SOCS3 upregulation. Moreover, JAK2-STAT3/NFκB signaling predominates the pathogenesis of VILI. Nevertheless, the interaction between SOCS3 and WNK4 in modulating VILI in obesity warrants further investigation.


Subject(s)
Obesity/complications , STAT3 Transcription Factor/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/metabolism , Ventilator-Induced Lung Injury/complications , Ventilator-Induced Lung Injury/metabolism , Animals , Biomarkers , Cytokines/metabolism , Diet, High-Fat , Disease Models, Animal , Disease Progression , Disease Susceptibility , Gene Expression Regulation , Gene Knockdown Techniques , Mice , Mice, Knockout , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Suppressor of Cytokine Signaling 3 Protein/genetics , Ventilator-Induced Lung Injury/etiology
7.
Front Pharmacol ; 12: 650573, 2021.
Article in English | MEDLINE | ID: mdl-34335242

ABSTRACT

Background: Poloxamer 188 (P188) possesses anti-inflammatory properties and can help to maintain plasma membrane function. P188 has been reported to exert beneficial effects in the treatment of various disorders. However, the effects of P188 in ischemia/reperfusion (IR)-induced acute lung injury have not been examined. Methods: We investigated the ability of P188 to attenuate IR-induced acute lung injury in rats and hypoxia/reoxygenation (HR) injury in murine epithelial cells. Isolated perfused rat lungs were exposed to 40 min ischemia followed by 60 min reperfusion to induce IR injury. Results: IR led to lung edema, increased pulmonary arterial pressure, promoted lung tissue inflammation and oxidative stress, and upregulated the levels of TNF-α, IL-6 and CINC-1, and increased Lactic dehydrogenase (LDH) activity in bronchoalveolar lavage fluid. IR also downregulated the levels of inhibitor of κB (IκB-α), upregulated nuclear factor (NF)-κB (NF-κB), and promoted apoptosis in lung tissues. P188 significantly suppressed all these effects. In vitro, P188 also exerted a similar effect in murine lung epithelial cells exposed to HR. Furthermore, P188 reduced the number of propidium iodide-positive cells, maintained cell membrane integrity, and enhanced cell membrane repair following HR. Conclusion: We conclude that P188 protects against lung IR injury by suppressing multiple signaling pathways and maintaining cell membrane integrity.

8.
Br J Neurosurg ; : 1-2, 2021 Aug 19.
Article in English | MEDLINE | ID: mdl-34410216

ABSTRACT

Cerebral air embolism (CAE) is considered as a rare complication during the routine medical procedures in the literature review. We reported a very rare complication of CAE after the percutaneous kyphoplasty (PKP) for the treatment of acute vertebral compression fracture.

9.
Front Immunol ; 12: 674316, 2021.
Article in English | MEDLINE | ID: mdl-34122432

ABSTRACT

Endoplasmic reticulum (ER) stress that disrupts ER function can occur in response to a wide variety of cellular stress factors leads to the accumulation of unfolded and misfolded proteins in the ER. Many studies have shown that ER stress amplified inflammatory reactions and was involved in various inflammatory diseases. However, little is known regarding the role of ER stress in hyperoxia-induced acute lung injury (HALI). This study investigated the influence of ER stress inhibitor, 4-phenyl butyric acid (4-PBA), in mice with HALI. Treatment with 4-PBA in the hyperoxia groups significantly prolonged the survival, decreased lung edema, and reduced the levels of inflammatory mediators, lactate dehydrogenase, and protein in bronchoalveolar lavage fluid, and increased claudin-4 protein expression in lung tissue. Moreover, 4-PBA reduced the ER stress-related protein expression, NF-κB activation, and apoptosis in the lung tissue. In in vitro study, 4-PBA also exerted a similar effect in hyperoxia-exposed mouse lung epithelial cells (MLE-12). However, when claudin-4 siRNA was administrated in mice and MLE-12 cells, the protective effect of 4-PBA was abrogated. These results suggested that 4-PBA protected against hyperoxia-induced ALI via enhancing claudin-4 expression.


Subject(s)
Acute Lung Injury/metabolism , Butylamines/pharmacology , Claudin-4/metabolism , Endoplasmic Reticulum Stress/drug effects , Acute Lung Injury/etiology , Animals , Hyperoxia/complications , Male , Mice , Mice, Inbred C57BL , Up-Regulation
10.
Front Immunol ; 12: 596376, 2021.
Article in English | MEDLINE | ID: mdl-33796096

ABSTRACT

Background: 2-Methoxyestradiol (2ME), a natural 17-ß estradiol metabolite, is a potent anti-inflammatory agent, but its effect on ischemia/reperfusion (IR)-induced acute lung inflammation remains unknown. Annexin A1 (AnxA1), a glucocorticoid-regulated protein, is effective at inhibiting neutrophil transendothelial migration by binding the formyl peptide receptors (FPRs). We aimed to investigate whether 2ME upregulates the expression of AnxA1 and protects against IR-induced lung damage. Methods: IR-mediated acute lung inflammation was induced by ischemia for 40 min followed by reperfusion for 60 min in an isolated, perfused rat lung model. The rat lungs were randomly treated with vehicle or 2ME, and the functional relevance of AnxA1 was determined using an anti-AnxA1 antibody or BOC2 (a pan-receptor antagonist of the FPR). In vitro, human primary alveolar epithelial cells (HPAECs) and rat neutrophils were pretreated with 2ME and an AnxA1 siRNA or anti-AnxA1 antibody and subjected to hypoxia-reoxygenation (HR). Results: 2ME significantly decreased all lung edema parameters, neutrophil infiltration, oxidative stress, proinflammatory cytokine production, lung cell apoptosis, tight junction protein disruption, and lung tissue injury in the IR-induced acute lung inflammation model. 2ME also increased the expression of the AnxA1 mRNA and protein and suppressed the activation of nuclear factor-κB (NF-κB). In vitro, 2ME attenuated HR-triggered NF-κB activation and interleukin-8 production in HPAECs, decreased transendothelial migration, tumor necrosis factor-α production, and increased apoptosis in neutrophils exposed to HR. These protective effects of 2ME were significantly abrogated by BOC2, the anti-AnxA1 antibody, or AnxA1 siRNA. Conclusions: 2ME ameliorates IR-induced acute lung inflammation by increasing AnxA1 expression. Based on these results, 2ME may be a promising agent for attenuating IR-induced lung injury.


Subject(s)
2-Methoxyestradiol/pharmacology , Annexin A1/immunology , Lung Diseases , Lung/immunology , Reperfusion Injury/immunology , Up-Regulation/drug effects , Animals , Lung Diseases/immunology , Lung Diseases/prevention & control , Male , Rats , Rats, Sprague-Dawley , Reperfusion Injury/prevention & control , Up-Regulation/immunology
11.
Int J Med Sci ; 18(6): 1363-1374, 2021.
Article in English | MEDLINE | ID: mdl-33628092

ABSTRACT

Introduction: Acute lung injury (ALI) has a great impact and a high mortality rate in intensive care units (ICUs). Excessive air may enter the lungs, causing pulmonary air embolism (AE)-induced ALI. Some invasive iatrogenic procedures cause pulmonary AE-induced ALI, with the presentation of severe inflammatory reactions, hypoxia, and pulmonary hypertension. Pulmonary surfactants are vital in the lungs to reduce the surface tension and inflammation. Nonionic surfactants (NIS) are a kind of surfactants without electric charge on their hydrophilic parts. Studies on NIS in AE-induced ALI are limited. We aimed to study the protective effects and mechanisms of NIS in AE-induced ALI. Materials and methods: Five different groups (n = 6 in each group) were created: sham, AE, AE + NIS pretreatment (0.5 mg/kg), AE + NIS pretreatment (1 mg/kg), and AE + post-AE NIS (1 mg/kg). AE-induced ALI was introduced by the infusion of air via the pulmonary artery. Aerosolized NIS were administered via tracheostomy. Results: Pulmonary AE-induced ALI showed destruction of the alveolar cell integrity with increased pulmonary microvascular permeability, pulmonary vascular resistance, pulmonary edema, and lung inflammation. The activation of nuclear factor-κB (NF-κB) increased the expression of pro-inflammatory cytokines, and sodium-potassium-chloride co-transporter isoform 1 (NKCC1). The pretreatment with NIS (1 mg/kg) prominently maintained the integrity of the epithelial lining and suppressed the expression of NF-κB, pro-inflammatory cytokines, and NKCC1, subsequently reducing AE-induced ALI. Conclusions: NIS maintained the integrity of the epithelial lining and suppressed the expression of NF-κB, pro-inflammatory cytokines, and NKCC1, thereby reducing hyperpermeability, pulmonary edema, and inflammation in ALI.


Subject(s)
Acute Lung Injury/prevention & control , Pulmonary Alveoli/drug effects , Pulmonary Embolism/drug therapy , Respiratory Mucosa/drug effects , Surface-Active Agents/administration & dosage , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Administration, Inhalation , Aerosols , Animals , Cytokines/metabolism , Disease Models, Animal , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Male , NF-kappa B/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Embolism/complications , Pulmonary Embolism/pathology , Rats , Respiratory Mucosa/metabolism , Respiratory Mucosa/ultrastructure , Signal Transduction/drug effects , Signal Transduction/immunology
12.
BMC Pulm Med ; 21(1): 58, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33588817

ABSTRACT

BACKGROUND: Hyperoxia downregulates the tight junction (TJ) proteins of the alveolar epithelium and leads to barrier dysfunction. Previous study has showed that STE20/SPS1-related proline/alanine-rich kinase (SPAK) interferes with the intestinal barrier function in mice. The aim of the present study is to explore the association between SPAK and barrier function in the alveolar epithelium after hyperoxic exposure. METHODS: Hyperoxic acute lung injury (HALI) was induced by exposing mice to > 99% oxygen for 64 h. The mice were randomly allotted into four groups comprising two control groups and two hyperoxic groups with and without SPAK knockout. Mouse alveolar MLE-12 cells were cultured in control and hyperoxic conditions with or without SPAK knockdown. Transepithelial electric resistance and transwell monolayer permeability were measured for each group. In-cell western assay was used to screen the possible mechanism of p-SPAK being induced by hyperoxia. RESULTS: Compared with the control group, SPAK knockout mice had a lower protein level in the bronchoalveolar lavage fluid in HALI, which was correlated with a lower extent of TJ disruption according to transmission electron microscopy. Hyperoxia down-regulated claudin-18 in the alveolar epithelium, which was alleviated in SPAK knockout mice. In MLE-12 cells, hyperoxia up-regulated phosphorylated-SPAK by reactive oxygen species (ROS), which was inhibited by indomethacin. Compared with the control group, SPAK knockdown MLE-12 cells had higher transepithelial electrical resistance and lower transwell monolayer permeability after hyperoxic exposure. The expression of claudin-18 was suppressed by hyperoxia, and down-regulation of SPAK restored the expression of claudin-18. The process of SPAK suppressing the expression of claudin-18 and impairing the barrier function was mediated by p38 mitogen-activated protein kinase (MAPK). CONCLUSIONS: Hyperoxia up-regulates the SPAK-p38 MAPK signal pathway by ROS, which disrupts the TJ of the alveolar epithelium by suppressing the expression of claudin-18. The down-regulation of SPAK attenuates this process and protects the alveolar epithelium against the barrier dysfunction induced by hyperoxia.


Subject(s)
Acute Lung Injury/metabolism , Alveolar Epithelial Cells/metabolism , Claudins/genetics , Hyperoxia/metabolism , Protein Serine-Threonine Kinases/genetics , Pulmonary Alveoli/metabolism , Tight Junctions/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Acute Lung Injury/pathology , Alveolar Epithelial Cells/ultrastructure , Animals , Bronchoalveolar Lavage Fluid/chemistry , Claudins/metabolism , Disease Models, Animal , Gene Expression Regulation , Gene Knockdown Techniques , Hyperoxia/pathology , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Permeability , Protein Serine-Threonine Kinases/metabolism , Pulmonary Alveoli/ultrastructure , Reactive Oxygen Species/metabolism , Signal Transduction , Tight Junctions/ultrastructure
13.
Inflammation ; 44(1): 57-67, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33089374

ABSTRACT

Excessive amounts of air can enter the lungs and cause air embolism (AE)-induced acute lung injury (ALI). Pulmonary AE can occur during diving, aviation, and iatrogenic invasive procedures. AE-induced lung injury presents with severe hypoxia, pulmonary hypertension, microvascular hyper-permeability, and severe inflammatory responses. Pulmonary AE-induced ALI is a serious complication resulting in significant morbidity and mortality. Surfactant is abundant in the lungs and its function is to lower surface tension. Earlier studies have explored the beneficial effects of surfactant in ALI; however, none have investigated the role of surfactant in pulmonary AE-induced ALI. Therefore, we conducted this study to determine the effects of surfactant in pulmonary AE-induced ALI. Isolated-perfused rat lungs were used as a model of pulmonary AE. The animals were divided into four groups (n = 6 per group): sham, air embolism (AE), AE + surfactant (0.5 mg/kg), and AE+ surfactant (1 mg/kg). Surfactant pretreatment was administered before the induction of pulmonary AE. Pulmonary AE was induced by the infusion of 0.7 cc air through a pulmonary artery catheter. After induction of air, pulmonary AE was presented with pulmonary edema, pulmonary microvascular hyper-permeability, and lung inflammation with neutrophilic sequestration. Activation of NF-κB was observed, along with increased expression of pro-inflammatory cytokines, and Na-K-Cl cotransporter isoform 1 (NKCC1). Surfactant suppressed the activation of NF-κB and decreased the expression of pro-inflammatory cytokines and NKCC1, thereby attenuating AE-induced lung injury. Therefore, AE-induced ALI presented with pulmonary edema, microvascular hyper-permeability, and lung inflammation. Surfactant suppressed the expressions of NF-κB, pro-inflammatory cytokines, and NKCC1, thereby attenuating AE-induced lung injury.


Subject(s)
Acute Lung Injury/drug therapy , Embolism, Air/drug therapy , NF-kappa B/antagonists & inhibitors , Solute Carrier Family 12, Member 2/biosynthesis , Surface-Active Agents/therapeutic use , Acute Lung Injury/genetics , Acute Lung Injury/metabolism , Animals , Embolism, Air/genetics , Embolism, Air/metabolism , Gene Expression Regulation , Male , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Solute Carrier Family 12, Member 2/genetics , Surface-Active Agents/pharmacology
14.
Interact Cardiovasc Thorac Surg ; 31(5): 718-726, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33051664

ABSTRACT

OBJECTIVES: An optimal ventilation strategy that causes as little mechanical stress and inflammation as possible is critical for patients undergoing pneumonectomy. The aim of this study was to determine whether adaptive support ventilation (ASV) can provide protective ventilation to the remaining lung after pneumonectomy with minimal mechanical stress and less inflammation than volume-control ventilation (VCV). METHODS: In this study, 15 pigs were randomly allocated to 3 groups (n = 5 for each group): the control group, the VCV group and the ASV group. After left pneumonectomy, the VCV group was treated with the volume-control set to 20 ml/kg, and the ASV group with the mode set to achieve 60% of the minute ventilation of 2 lungs. RESULTS: The ASV group had lower alveolar strain than the VCV group. The ASV group exhibited less lung injury and greater alveolar fluid clearance than the VCV group (13.3% vs -17.8%; P ≤ 0.018). Ventilator-induced lung injury was associated with changes in the cytokine levels in the exhaled breath condensate, differential changes in plasma and changes in the cytokines in the bronchoalveolar lavage fluid. Expression of 3 microRNAs (miR449b-3p, P ≤ 0.001; miR451-5p, P = 0.027; and miR144-5p, P = 0.008) was increased in the VCV group compared with the ASV group. CONCLUSIONS: The ASV mode was capable of supporting rapid, shallow breathing patterns to exert lung-protective effects in a porcine postpneumonectomy model. Further investigation of microRNAs as biomarkers of ventilator-induced lung injury is warranted.


Subject(s)
Acute Lung Injury , Lung , Pneumonectomy , Respiration, Artificial , Respiratory Distress Syndrome , Animals , Male , Acute Lung Injury/complications , Acute Lung Injury/physiopathology , Acute Lung Injury/therapy , Disease Models, Animal , Lung/physiopathology , Pneumonectomy/adverse effects , Respiration, Artificial/methods , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/therapy , Swine
15.
Int J Mol Sci ; 21(13)2020 Jul 07.
Article in English | MEDLINE | ID: mdl-32645929

ABSTRACT

Acute lung injury (ALI) is characterized by severe hypoxemia and has significantly high mortality rates. Acute hyperglycemia occurs in patients with conditions such as sepsis or trauma, among others, and it results in aggravated inflammation and induces damage in patients with ALI. Regulation of alveolar fluid is essential for the development and resolution of pulmonary edema in lung injury. Pulmonary sodium-potassium-chloride co-transporter 1 (NKCC1) regulates the net influx of ions and water into alveolar cells. The activation of with-no-lysine kinase 4 (WNK4), STE20/SPS1-related proline/alanine rich kinase (SPAK) and the NKCC1 pathway lead to an increase in the expression of NKCC1 and aggravation of ALI. Moreover, hyperglycemia is known to induce NKCC1 expression via the activation of the serum-glucocorticoid kinase 1 (SGK1)-NKCC1 pathway. We aim to evaluate the influence of acute hyperglycemia on the SGK1-NKCC1 pathway in ALI. ALI was induced using a high tidal volume for four hours in a rat model. Acute hyperglycemia was induced by injection with 0.5 mL of 40% glucose solution followed by continuous infusion at 2 mL/h. The animals were divided into sham, sham+ hyperglycemia, ALI, ALI + hyperglycemia, ALI + inhaled bumetanide (NKCC1 inhibitor) pretreatment, ALI + hyperglycemia + inhalational bumetanide pretreatment, and ALI + hyperglycemia + post-ALI inhalational bumetanide groups. Severe lung injury along with pulmonary edema, alveolar protein leakage, and lung inflammation was observed in ALI with hyperglycemia than in ALI without hyperglycemia. This was concurrent with the higher expression of pro-inflammatory cytokines, infiltration of neutrophils and alveolar macrophages (AM) 1, and NKCC1 expression. Inhalational NKCC1 inhibitor significantly inhibited the SGK1-NKCC1, and WNK4-SPAK-NKCC1 pathways. Additionally, it reduced pulmonary edema, inflammation, levels of pro-inflammatory cytokines, neutrophils and AM1 and increased AM2. Therefore, acute hyperglycemia aggravates lung injury via the further activation of the SGK1-NKCC1 pathway. The NKCC1 inhibitor can effectively attenuate lung injury aggravated by acute hyperglycemia.


Subject(s)
Acute Lung Injury/metabolism , Hyperglycemia/metabolism , Immediate-Early Proteins/metabolism , Lung/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Solute Carrier Family 12, Member 2/metabolism , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Animals , Bumetanide/pharmacology , Hyperglycemia/drug therapy , Lung/drug effects , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Male , Pneumonia/drug therapy , Pneumonia/metabolism , Pulmonary Edema/drug therapy , Pulmonary Edema/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
16.
Respir Res ; 21(1): 65, 2020 Mar 06.
Article in English | MEDLINE | ID: mdl-32143642

ABSTRACT

BACKGROUND: It is well known that ventilation with high volume or pressure may damage healthy lungs or worsen injured lungs. Melatonin has been reported to be effective in animal models of acute lung injury. Melatonin exerts its beneficial effects by acting as a direct antioxidant and via melatonin receptor activation. However, it is not clear whether melatonin receptor agonist has a protective effect in ventilator-induced lung injury (VILI). Therefore, in this study, we determined whether ramelteon (a melatonin receptor agonist) can attenuate VILI and explore the possible mechanism for protection. METHODS: VILI was induced by high tidal volume ventilation in a rat model. The rats were randomly allotted into the following groups: control, control+melatonin, control+ramelteon, control+luzindole, VILI, VILI+luzindole, VILI + melatonin, VILI + melatonin + luzindole (melatonin receptor antagonist), VILI + ramelteon, and VILI + ramelteon + luzindole (n = 6 per group). The role of interleukin-10 (IL-10) in the melatonin- or ramelteon-mediated protection against VILI was also investigated. RESULTS: Ramelteon treatment markedly reduced lung edema, serum malondialdehyde levels, the concentration of inflammatory cytokines in bronchoalveolar lavage fluid (BALF), NF-κB activation, iNOS levels, and apoptosis in the lung tissue. Additionally, ramelteon treatment significantly increased heat shock protein 70 expression in the lung tissue and IL-10 levels in BALF. The protective effect of ramelteon was mitigated by the administration of luzindole or an anti-IL-10 antibody. CONCLUSIONS: Our results suggest that a melatonin receptor agonist has a protective effect against VILI, and its protective mechanism is based on the upregulation of IL-10 production.


Subject(s)
Indenes/therapeutic use , Interleukin-10/biosynthesis , Receptors, Melatonin/agonists , Up-Regulation/drug effects , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/prevention & control , Animals , Indenes/pharmacology , Male , Rats , Rats, Sprague-Dawley , Up-Regulation/physiology
17.
Biochem Pharmacol ; 171: 113738, 2020 01.
Article in English | MEDLINE | ID: mdl-31786261

ABSTRACT

Dysregulation of alveolar macrophage activation has been recognized as the major mechanism in the pathogenesis of acute lung injury. The aim of the present study was to investigate the role of NKCC1 regulating mechanism in modulating macrophage activation. Knockout (SPAK-/- and WNK4-/-) and knockin (WNK4D561A/+) mice were used in this study. LPS induced expression of p-NKCC1 and activation of NFκB in the primary culture of alveolar macrophages. WNK4 or SPAK knockout suppressed p-NKCC1 expression and inflammation cascade activation, whereas WNK4 knockin enhanced these responses. Intrapulmonary administration of LPS induced in vivo expression and phosphorylation of NKCC1 in alveolar inflammation cells and caused a shift in the cell population from macrophage to neutrophil predominance. WNK4 or SPAK knockout attenuated the LPS-induced alveolar cell-population shifting, macrophage NKCC1 phosphorylation, and acute lung injury, whereas WNK4 knockin augmented the inflammatory response. In summary, our results demonstrated the presence of NKCC1 in alveolar macrophage, which is inducible by lipopolysaccharide. Our results also showed showed that the WNK4-SPAK-NKCC1 cascade plays an important role in modulating macrophage activation to regulate LPS-induced lung inflammation and lung injury.


Subject(s)
NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Solute Carrier Family 12, Member 2/metabolism , Animals , Cells, Cultured , Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung Injury/chemically induced , Lung Injury/genetics , Lung Injury/metabolism , Macrophages, Alveolar/cytology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-kappa B/genetics , Phosphorylation/drug effects , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/metabolism , Protein Serine-Threonine Kinases/genetics , Solute Carrier Family 12, Member 2/genetics
18.
Int J Mol Sci ; 20(24)2019 Dec 13.
Article in English | MEDLINE | ID: mdl-31847115

ABSTRACT

Various animal studies have shown beneficial effects of hypercapnia in lung injury. However, in patients with acute respiratory distress syndrome (ARDS), there is controversial information regarding the effect of hypercapnia on outcomes. The duration of carbon dioxide inhalation may be the key to the protective effect of hypercapnia. We investigated the effect of pre-treatment with inhaled carbon dioxide on lipopolysaccharide (LPS)-induced lung injury in mice. C57BL/6 mice were randomly divided into a control group or an LPS group. Each LPS group received intratracheal LPS (2 mg/kg); the LPS groups were exposed to hypercapnia (5% carbon dioxide) for 10 min or 60 min before LPS. Bronchoalveolar lavage fluid (BALF) and lung tissues were collected to evaluate the degree of lung injury. LPS significantly increased the ratio of lung weight to body weight; concentrations of BALF protein, tumor necrosis factor-α, and CXCL2; protein carbonyls; neutrophil infiltration; and lung injury score. LPS induced the degradation of the inhibitor of nuclear factor-κB-α (IκB-α) and nuclear translocation of NF-κB. LPS increased the surface protein expression of toll-like receptor 4 (TLR4). Pre-treatment with inhaled carbon dioxide for 10 min, but not for 60 min, inhibited LPS-induced pulmonary edema, inflammation, oxidative stress, lung injury, and TLR4 surface expression, and, accordingly, reduced NF-κB signaling. In summary, our data demonstrated that pre-treatment with 10-min carbon dioxide inhalation can ameliorate LPS-induced lung injury. The protective effect may be associated with down-regulation of the surface expression of TLR4 in the lungs.


Subject(s)
Acute Lung Injury , Carbon Dioxide/pharmacology , Down-Regulation/drug effects , Lipopolysaccharides/toxicity , Respiratory Distress Syndrome , Signal Transduction/drug effects , Toll-Like Receptor 4/biosynthesis , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Male , Mice , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology
19.
Int J Mol Sci ; 20(23)2019 Nov 21.
Article in English | MEDLINE | ID: mdl-31766467

ABSTRACT

Adaptive support ventilation (ASV) is a closed-loop ventilation, which can make automatic adjustments in tidal volume (VT) and respiratory rate based on the minimal work of breathing. The purpose of this research was to study whether ASV can provide a protective ventilation pattern to decrease the risk of ventilator-induced lung injury in patients of acute respiratory distress syndrome (ARDS). In the clinical study, 15 ARDS patients were randomly allocated to an ASV group or a pressure-control ventilation (PCV) group. There was no significant difference in the mortality rate and respiratory parameters between these two groups, suggesting the feasible use of ASV in ARDS. In animal experiments of 18 piglets, the ASV group had a lower alveolar strain compared with the volume-control ventilation (VCV) group. The ASV group exhibited less lung injury and greater alveolar fluid clearance compared with the VCV group. Tissue analysis showed lower expression of matrix metalloproteinase 9 and higher expression of claudin-4 and occludin in the ASV group than in the VCV group. In conclusion, the ASV mode is capable of providing ventilation pattern fitting into the lung-protecting strategy; this study suggests that ASV mode may effectively reduce the risk or severity of ventilator-associated lung injury in animal models.


Subject(s)
Lung/physiopathology , Respiration, Artificial/methods , Respiratory Distress Syndrome/therapy , Tidal Volume/physiology , Ventilator-Induced Lung Injury/therapy , Adult , Aged , Aged, 80 and over , Animals , Claudin-4/metabolism , Female , Humans , Lung/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Middle Aged , Occludin/metabolism , Respiration , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/physiopathology , Swine , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/physiopathology
20.
BMJ Open ; 9(11): e031135, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31740467

ABSTRACT

OBJECTIVES: To identify the risk factors related to the prognosis of carbon monoxide (CO)-poisoned patients in the hospital. DESIGN: Retrospective observational study. SETTING: Tri-Service General Hospital, Taiwan. METHODS: We conducted a review of the medical records of 669 CO-poisoned patients, who were admitted to the Department of Emergency, Tri-Service General Hospital, Taiwan, from 2009 to 2014. Demographic, clinical and laboratory data were collected for analysis. In the study, the end points for poor outcome were patients who either still had sequelae, were bedridden or died after treatment. The independent t-test, χ2 test and binary logistic regression were used to identify the association between the prognostic factors and the outcomes. RESULTS: The logistic regression analysis confirmed that the Glasgow Coma Scale (GCS) score (p=0.008) and blood urea nitrogen (BUN) (p=0.002) were related to poor outcomes. Furthermore, the receiver operating characteristic (ROC) curve showed that the cut-off point of intubation days was 1.5 days (area under the ROC curve [AUC]=0.793) for all patients and 2.5 days (AUC=0.817) for patients with intubation when predicting poor outcomes. CONCLUSION: We identified the factors that most strongly predict the prognosis of CO poisoning, including the GCS score, serum BUN and intubation days. Moreover, the number of hyperbaric oxygen treatments seems to have impact of the outcome.


Subject(s)
Carbon Monoxide Poisoning/mortality , Adult , Carbon Monoxide Poisoning/therapy , Comorbidity , Female , Humans , Hyperbaric Oxygenation/adverse effects , Hyperbaric Oxygenation/statistics & numerical data , Male , Middle Aged , ROC Curve , Retrospective Studies , Risk Factors , Suicide, Attempted/statistics & numerical data , Taiwan/epidemiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...