Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 807
Filter
1.
J Agric Food Chem ; 72(38): 20944-20958, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39259217

ABSTRACT

Radiation-induced lung injury (RILI) is a prevalent complication of thoracic tumor radiotherapy and accidental radiation exposure. Pyrroloquinoline quinone (PQQ), a novel vitamin B, plays a crucial role in delaying aging, antioxidation, anti-inflammation, and antiapoptosis. This study aims to investigate the protective effect and mechanisms of PQQ against RILI. C57BL/6 mice were exposed to a 20 Gy dose of X-ray radiation on the entire thorax with or without daily oral administration of PQQ for 2 weeks. PQQ effectively mitigated radiation-induced lung tissue damage, inflammation, oxidative stress, and epithelial cell apoptosis. Additionally, PQQ significantly inhibited oxidative stress and mitochondrial damage in MLE-12 cells. Mechanistically, PQQ upregulated the mRNA and protein levels of MOTS-c in irradiated lung tissue and MLE-12 cells. Knockdown of MOTS-c by siRNA substantially attenuated the protective effects of PQQ on oxidative stress, inflammation, and apoptosis. In conclusion, PQQ alleviates RILI by preserving mitochondrial function through a MOTS-c-dependent mechanism, suggesting that PQQ may serve as a promising nutraceutical intervention against RILI.


Subject(s)
Apoptosis , Lung Injury , Mice, Inbred C57BL , Mitochondria , Oxidative Stress , PQQ Cofactor , Animals , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/radiation effects , PQQ Cofactor/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Lung Injury/metabolism , Lung Injury/etiology , Lung Injury/genetics , Lung Injury/prevention & control , Lung Injury/drug therapy , Humans , Apoptosis/drug effects , Male , Radiation Injuries/metabolism , Radiation Injuries/genetics , Radiation Injuries/drug therapy , Radiation Injuries/prevention & control , Lung/radiation effects , Lung/metabolism , Lung/drug effects
2.
Nat Commun ; 15(1): 8226, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300060

ABSTRACT

Hemolysis drives susceptibility to lung injury and predicts poor outcomes in diseases, such as malaria and sickle cell disease (SCD). However, the underlying pathological mechanism remains elusive. Here, we report that major facilitator superfamily domain containing 7 C (MFSD7C) protects the lung from hemolytic-induced damage by preventing ferroptosis. Mechanistically, MFSD7C deficiency in HuLEC-5A cells leads to mitochondrial dysfunction, lipid remodeling and dysregulation of ACSL4 and GPX4, thereby enhancing lipid peroxidation and promoting ferroptosis. Furthermore, systemic administration of MFSD7C mRNA-loaded nanoparticles effectively prevents lung injury in hemolytic mice, such as HbSS-Townes mice and PHZ-challenged 7 C-/- mice. These findings present the detailed link between hemolytic complications and ferroptosis, providing potential therapeutic targets for patients with hemolytic disorders.


Subject(s)
Ferroptosis , Hemolysis , Mice, Knockout , Phospholipid Hydroperoxide Glutathione Peroxidase , Animals , Female , Humans , Male , Mice , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/genetics , Coenzyme A Ligases/metabolism , Coenzyme A Ligases/genetics , Disease Models, Animal , Ferroptosis/drug effects , Ferroptosis/genetics , Hemolysis/drug effects , Lipid Peroxidation/drug effects , Lung/pathology , Lung/metabolism , Lung Injury/metabolism , Lung Injury/pathology , Lung Injury/prevention & control , Lung Injury/genetics , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/drug effects , Nanoparticles/chemistry , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics
3.
Tomography ; 10(9): 1342-1353, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39330747

ABSTRACT

BACKGROUND: Radiation-induced lung injury (RILI), a serious side effect of thoracic radiotherapy, can lead to acute radiation pneumonitis (RP) and chronic pulmonary fibrosis (PF). Despite various interventions, no effective protocol exists to prevent pneumonitis. Oxytocin (OT), known for its anti-inflammatory, antiapoptotic, and antioxidant properties, has not been explored for its potential in mitigating RILI. MATERIALS AND METHODS: This study involved 24 female Wistar albino rats, divided into three groups: control group, radiation (RAD) + saline, and RAD + OT. The RAD groups received 18 Gy of whole-thorax irradiation. The RAD + OT group was treated with OT (0.1 mg/kg/day) intraperitoneally for 16 weeks. Computerizing tomography (CT) imaging and histopathological, biochemical, and blood gas analyses were performed to assess lung tissue damage and inflammation. RESULTS: Histopathological examination showed significant reduction in alveolar wall thickening, inflammation, and vascular changes in the RAD + OT group compared to the RAD + saline group. Biochemical analysis revealed decreased levels of TGF-beta, VEGF, and PDGF, and increased BMP-7 and prostacyclin in the RAD + oxytocin group (p < 0.05). Morphometric analysis indicated significant reductions in fibrosis, edema, and immune cell infiltration. CT imaging demonstrated near-normal lung parenchyma density in the RAD + oxytocin group (p < 0.001). CONCLUSION: Oxytocin administration significantly mitigates radiation-induced pneumonitis in rats, implying that is has potential as a therapeutic agent for preventing and treating RILI.


Subject(s)
Oxytocin , Rats, Wistar , Animals , Oxytocin/pharmacology , Oxytocin/therapeutic use , Female , Rats , Tomography, X-Ray Computed/methods , Lung/radiation effects , Lung/pathology , Lung/diagnostic imaging , Radiation Pneumonitis/pathology , Radiation Pneumonitis/drug therapy , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/diagnostic imaging , Lung Injury/etiology , Lung Injury/diagnostic imaging , Lung Injury/pathology , Lung Injury/prevention & control , Radiation-Protective Agents/pharmacology , Radiation-Protective Agents/therapeutic use
4.
Int J Immunopathol Pharmacol ; 38: 3946320241272642, 2024.
Article in English | MEDLINE | ID: mdl-39096175

ABSTRACT

Prolonged exposure to different occupational or environmental toxicants triggered oxidative stress and inflammatory reactions mediated lung damage. This study was designed to explore the influence and protective impact of flavone on lung injury in rats intoxicated with nicotine (NIC) and exposed to radiation (IR). Forty rats were divided into four groups; group I control, group II flavone; rats were administered with flavone (25 mg/kg/day), group III NIC + IR; rats were injected intraperitoneally with NIC (1 mg/kg/day) and exposed to γ-IR (3.5 Gy once/week for 2 weeks) while group IV NIC + IR + flavone; rats were injected with NIC, exposed to IR and administered with flavone. Redox status parameters and histopathological changes in lung tissue were evaluated. Nuclear factor-kappa B (NF-κB), forkhead box O-class1 (FoxO1) and nucleotide-binding domain- (NOD-) like receptor pyrin domain-containing-3 (NLRP3) gene expression were measured in lung tissues. Moreover, nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and phosphatidylinositol three kinase (PI3K) were measured using ELISA kits. Our data demonstrates, for the first time, that flavone protects the lung from NIC/IR-associated cytotoxicity, by attenuating the disrupted redox status and aggravating the antioxidant defence mechanism via activation of the PI3K/Nrf2. Moreover, flavone alleviates pulmonary inflammation by inhibiting the inflammatory signaling pathway FOXO1/NF-κB/NLRP3- Inflammasome. Collectively, the obtained results exhibited a notable efficiency of flavone in alleviating lung injury induced by NIC and IR via modulating PI3K/Nrf2 and FoxO1/NLRP3 Inflammasome.


Subject(s)
Flavones , Inflammasomes , Lung Injury , Nicotine , Animals , Male , Rats , Flavones/pharmacology , Forkhead Box Protein O1 , Gamma Rays , Inflammasomes/metabolism , Inflammasomes/drug effects , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung/radiation effects , Lung Injury/metabolism , Lung Injury/prevention & control , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Nicotine/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Rats, Wistar , Signal Transduction/drug effects
5.
Sheng Li Xue Bao ; 76(4): 517-525, 2024 Aug 25.
Article in Chinese | MEDLINE | ID: mdl-39192785

ABSTRACT

The aim of this study was to investigate whether the protective effect of 2-deoxyglucose (2-DG) on lung ischemia/reperfusion (I/R) injury is mediated by inhibiting nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3)-mediated pyroptosis in rats. Male Sprague-Dawley rats were randomly divided into control group, 2-DG group, lung I/R injury group (I/R group) and 2-DG+I/R group. 2-DG (0.7 g/kg) was intraperitoneally injected 1 h prior to lung ischemia. The tissue structure was measured under light microscope. Lung injury parameters were detected. The contents of malondialdehyde (MDA), myeloperoxidase (MPO) and lactate were determined by commercially available kits. ELISA was used to detect the levels of IL-1ß and IL-18. Western blot, qRT-PCR and immunofluorescence staining were used to measure the expression changes of glycolysis and pyroptosis related indicators. The results showed that there was no significant difference in the parameters between the control group and the 2-DG group. However, the lung injury parameters, oxidative stress response, lactic acid content, IL-1ß, and IL-18 levels were significantly increased in the I/R group. The protein expression levels of glycolysis and pyroptosis related indicators including hexokinase 2 (HK2), pyruvate kinase 2 (PKM2), NLRP3, Gasdermin superfamily member GSDMD-N, cleaved-Caspase1, cleaved-IL-1ß and cleaved-IL-18, and the gene expression levels of HK2, PKM2 and NLRP3 were markedly up-regulated in the I/R group compared with those in the control group. The expression of HK2 and NLRP3 was also increased detected by immunofluorescence staining. Compared with the I/R group, the 2-DG+I/R group exhibited significantly improved alveolar structure and inflammatory infiltration, reduced lung injury parameters, and decreased expression of glycolysis and pyroptosis related indicators. These results suggest that 2-DG protects against lung I/R injury possibly by inhibiting NLRP3-mediated pyroptosis in rats.


Subject(s)
Deoxyglucose , Lung , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Rats, Sprague-Dawley , Reperfusion Injury , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Male , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Rats , Lung/metabolism , Lung/pathology , Deoxyglucose/pharmacology , Interleukin-1beta/metabolism , Interleukin-18/metabolism , Lung Injury/metabolism , Lung Injury/prevention & control , Lung Injury/etiology , Oxidative Stress
6.
Immun Inflamm Dis ; 12(8): e70001, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39172009

ABSTRACT

BACKGROUND: The inhalation of paraquat (PQ), one of the most widely used herbicides in the world, can result in lung injury. Curcuma longa (Cl) has long history in traditional and folk medicine for the treatment of a wide range of disorders including respiratory diseases. AIM: The aim of the present work was to evaluate the preventive effect of Cl on inhaled PQ-induced lung injury in rats. METHODS: Male Wistar rats were divided into 8 groups (n = 7), one group exposed to saline (control) and other groups exposed to PQ aerosol. Saline (PQ), Cl extract, (two doses), curcumin (Cu), pioglitazone (Pio), and the combination of Cl-L + Pio and dexamethasone (Dex) were administered during the exposure period to PQ. Total and differential white blood cell (WBC) counts, oxidant and antioxidant indicators in the bronchoalveolar lavage (BALF), interleukin (IL)-10, and tumor necrosis alpha (TNF-α) levels in the lung tissues, lung histologic lesions score, and air way responsiveness to methacholine were evaluated. RESULTS: WBC counts (Total and differential), malondialdehyde level, tracheal responsiveness (TR), IL-10, TNF-α and histopathological changes of the lung were markedly elevated but total thiol content and the activities of catalase and superoxide dismutase were decreased in the BALF in the PQ group. Both doses of Cl, Cu, Pio, Cl-L + Pio, and Dex markedly improved all measured variables in comparison with the PQ group. CONCLUSION: CI, Pio, and Cl-L + Pio improved PQ-induced lung inflammation and oxidative damage comparable with the effects of Dex.


Subject(s)
Curcuma , PPAR gamma , Paraquat , Pioglitazone , Plant Extracts , Rats, Wistar , Animals , Pioglitazone/pharmacology , Pioglitazone/therapeutic use , Paraquat/toxicity , Male , Rats , Curcuma/chemistry , PPAR gamma/agonists , PPAR gamma/metabolism , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Lung/pathology , Lung/drug effects , Lung/metabolism , Lung Injury/chemically induced , Lung Injury/prevention & control , Lung Injury/drug therapy , Lung Injury/pathology , Lung Injury/metabolism , Dexamethasone/pharmacology , Bronchoalveolar Lavage Fluid/cytology , Oxidative Stress/drug effects , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use , Antioxidants/pharmacology , Curcumin/pharmacology , Curcumin/therapeutic use
7.
Biochem Pharmacol ; 227: 116418, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38996928

ABSTRACT

Ovarian tumor domain-containing protease 1 (OTUD1) is a critical negative regulator that promotes innate immune homeostasis and is extensively involved in the pathogenesis of sepsis. In this study, we performed a powerful integration of multiomics analysis and an experimental mechanistic investigation to elucidate the immunoregulatory role of OTUD1 in sepsis at the clinical, animal and cellular levels. Our study revealed the upregulation of OTUD1 expression and the related distinctive alterations observed via multiomics profiling in clinical and experimental sepsis. Importantly, in vivo and in vitro, OTUD1 was shown to negatively regulate inflammatory responses and play a protective role in sepsis-induced pathological lung injury by mechanistically inhibiting the activation of the transforming growth factor-beta-activated kinase 1 (TAK1)-mediated mitogen-activated protein kinase (MAPK) and nuclear factor kappa-B (NF-κB) signaling pathways in the present study. Subsequently, we probed the molecular mechanisms underlying OTUD1's regulation of NF-κB and MAPK pathways by pinpointing the target proteins that OTUD1 can deubiquitinate. Drawing upon prior research conducted in our laboratory, it has been demonstrated that tumor necrosis factor-α-induced protein 8-like 2 (TIPE2) performs a protective function in septic lung injury and septic encephalopathy by suppressing the NF-κB and MAPK pathways. Hence, we hypothesized that TIPE2 might be a target protein of OTUD1. Additional experiments, including Co-IP, immunofluorescence co-localization, and Western blotting, revealed that OTUD1 indeed has the ability to deubiquitinate TIPE2. In summary, OTUD1 holds potential as an immunoregulatory and inflammatory checkpoint agent, and could serve as a promising therapeutic target for sepsis-induced lung injury.


Subject(s)
Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase Kinases , Mice, Inbred C57BL , NF-kappa B , Sepsis , Ubiquitin-Specific Proteases , Animals , Humans , Male , Mice , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Lung Injury/metabolism , Lung Injury/etiology , Lung Injury/prevention & control , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Signaling System/physiology , NF-kappa B/metabolism , Sepsis/metabolism , Signal Transduction/physiology , Ubiquitin-Specific Proteases/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitination
8.
J Physiol Pharmacol ; 75(3)2024 Jun.
Article in English | MEDLINE | ID: mdl-39042394

ABSTRACT

Bronchopulmonary dysplasia (BPD) is a common serious complication of premature babies. No effective means control it. Hyperoxia damage is one of the important mechanisms of BPD. The reaserach confirmed pyroptosis existed in BPD. Dexmedetomidine is a new, high-specific α2 receptor agonist. Previous research foundation found that dexmedetomidine has a protective effect on BPD. To investigate how dexmedetomidine improves hyperoxic lung injury in neonatal mice by regulating pyroptosis. Neonatal rats were randomly divided into four groups: normal control group, hyperoxic injury group, air plus dexmedetomidine group, and hyperoxia plus dexmedetomidine group. After seven days the lungs of rats in each group were extracted, and the wet-to-dry weight ratio of the lung was measured. The lung injury in rats was observed using hematoxylin-eosin staining. Additionally, the expression and localization of nucleotide-binding oligomerization domain-like receptor thermal protein domain associated protein 3 (NLRP3), apoptosis-associated speck-like protein (ASC), and gasdermin D (GSDMD) proteins were examined in the lungs of rats using immunofluorescence staining. The mRNA levels of NLRP3, ASC, caspase-1, and interleukin 18 (IL-18) in the lungs of rats were determined using real-time PCR. Moreover, the protein levels of NLRP3, ASC, caspase-1/cleaved caspase-1, interleukin 1beta (IL-1ß), IL-18, and tunor necrosis factor alpha (TNF-α) were detected in lungs of rats using Western blot. The extent of mitochondrial damage in lung tissues of each group was observed by transmission electron microscopy. The lung tissue injury of the neonatal rats was significantly improved in the hyperoxia plus dexmedetomidine group compared to the hyperoxic injury group. Furthermore, the expressions of pyroptosis-related proteins such as NLRP3, ASC, cleaved-caspase-1, and GSDMD were significantly decreased, along with the expressions of inflammatory factors in lung tissues. By inhibiting the NLRP3/caspase-1/GSDMD pyroptosis pathway, dexmedetomidine reduces the activation and release of inflammatory factors and provides a protective effect against hyperoxic lung injury in neonatal mice.


Subject(s)
Animals, Newborn , Dexmedetomidine , Hyperoxia , Lung Injury , Lung , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Rats, Sprague-Dawley , Animals , Dexmedetomidine/pharmacology , Dexmedetomidine/therapeutic use , Hyperoxia/metabolism , Hyperoxia/complications , Hyperoxia/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Lung/drug effects , Lung/pathology , Lung/metabolism , Pyroptosis/drug effects , Lung Injury/metabolism , Lung Injury/prevention & control , Lung Injury/pathology , Lung Injury/drug therapy , Rats , Phosphate-Binding Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/metabolism , Interleukin-18/metabolism , Adrenergic alpha-2 Receptor Agonists/pharmacology , Adrenergic alpha-2 Receptor Agonists/therapeutic use , Male , Gasdermins
9.
Immunopharmacol Immunotoxicol ; 46(5): 594-603, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39048516

ABSTRACT

BACKGROUND: Inflammation and oxidative stress are key players in lung injury stemming from cardiac ischemia (LISCI). Cannabidiol (CBD) demonstrates tissue-protective properties through its antioxidant, anti-inflammatory, and anti-apoptotic characteristics. This study aims to assess the preventive (p-CBD) and therapeutic (t-CBD) effects of CBD on LISCI. METHODS: Forty male Wistar Albino rats were divided into four groups: control (CON), LISCI, p-CBD, and t-CBD. The left anterior descending coronary artery was ligated for 30 min of ischemia followed by 30 min of reperfusion. Lung tissues were then extracted for histopathological, immunohistochemical, genetic, and biochemical analyses. RESULTS: Histopathologically, marked hyperemia, increased septal tissue thickness, and inflammatory cell infiltrations were observed in the lung tissues of the LISCI group. Spectrophotometrically, total oxidant status and oxidative stress index levels were elevated, while total antioxidant status levels were decreased. Immunohistochemically, expressions of cyclooxygenase-1 (COX1), granulocyte colony-stimulating factor (GCSF), interleukin-6 (IL6) were increased. In genetic analyses, PERK and CHOP expressions were increased, whereas Nuclear factor erythroid 2-related factor 2 (NRF2) and B-cell leukemia/lymphoma 2 protein (BCL2) expressions were decreased. These parameters were alleviated by both prophylactic and therapeutic CBD treatment protocols. CONCLUSION: In LISCI-induced damage, both endoplasmic reticulum and mitochondrial stress, along with oxidative and inflammatory markers, were triggered, resulting in lung cell damage. However, both p-CBD and t-CBD treatments effectively reversed these mechanisms, normalizing all histopathological, biochemical, and PCR parameters.


Subject(s)
Cannabidiol , Lung Injury , Myocardial Ischemia , NF-E2-Related Factor 2 , Proto-Oncogene Proteins c-bcl-2 , Rats, Wistar , Transcription Factor CHOP , Animals , Cannabidiol/pharmacology , Male , Rats , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription Factor CHOP/metabolism , Lung Injury/prevention & control , Lung Injury/drug therapy , Lung Injury/metabolism , Lung Injury/pathology , Myocardial Ischemia/drug therapy , Myocardial Ischemia/pathology , Myocardial Ischemia/metabolism , Myocardial Ischemia/prevention & control , eIF-2 Kinase/metabolism , Disease Models, Animal , Signal Transduction/drug effects , Oxidative Stress/drug effects
10.
Sci Rep ; 14(1): 14231, 2024 06 20.
Article in English | MEDLINE | ID: mdl-38902260

ABSTRACT

Butorphanol is widely used as an anesthetic drug, whether butorphanol could reduce organ injury and protecting lung tissue is unknown. This study explored the effects of butorphanol on ALI and investigated its underlying mechanisms. We established a "two-hit" rat model and "two-hit" cell model to prove our hypothesis. Rats were divided into four groups [control, "two-hit" (OA + LPS), "two-hit" + butorphanol (4 mg/kg and 8 mg/kg) (OA + LPS + B1 and OA + LPS + B2)]. RPMVE cells were divided into four groups [control, "two-hit" (OA + LPS), "two-hit" + butorphanol (4 µM and 8 µM) (OA + LPS + 4 µM and OA + LPS + 8 µM)]. Inflammatory injury was assessed by the histopathology and W/D ratio, inflammatory cytokines, and arterial blood gas analysis. Apoptosis was assessed by Western blotting and flow cytometry. The effect of NF-κB p65 was detected by ELISA. Butorphanol could relieve the "two-hit" induced lung injury, the expression of TNF, IL-1ß, IL-6, and improve lung ventilation. In addition, butorphanol decreased Bax and cleaved caspase-3, increased an antiapoptotic protein (Bcl-2), and inhibited the "two-hit" cell apoptosis ratio. Moreover, butorphanol suppressed NF-κB p65 activity in rat lung injury. Our research showed that butorphanol may attenuate "two-hit"-induced lung injury by regulating the activity of NF-κB p65, which may supply more evidence for ALI treatment.


Subject(s)
Acute Lung Injury , Apoptosis , Butorphanol , Inflammation , Animals , Butorphanol/pharmacology , Apoptosis/drug effects , Rats , Male , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/drug therapy , Acute Lung Injury/prevention & control , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Transcription Factor RelA/metabolism , Lipopolysaccharides , Rats, Sprague-Dawley , Lung Injury/chemically induced , Lung Injury/drug therapy , Lung Injury/metabolism , Lung Injury/pathology , Lung Injury/prevention & control , Disease Models, Animal , Cytokines/metabolism , Lung/pathology , Lung/drug effects , Lung/metabolism
11.
Am J Nurs ; 124(7): 28-34, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38837249

ABSTRACT

ABSTRACT: Using a blind insertion technique to insert small-bore feeding tubes can result in inadvertent placement in the lungs, leading to lung perforation and even mortality. In a Magnet-designated, 500-bed, level 2 trauma center, two serious patient safety events occurred in a four-week period due to nurses blindly inserting a small-bore feeding tube. A patient safety event review team convened and conducted an assessment of reported small-bore feeding tube insertion events that occurred between March 2019 and July 2021. The review revealed six lung perforations over this two-year period. These events prompted the creation of a multidisciplinary team to evaluate alternative small-bore feeding tube insertion practices. The team reviewed the literature and evaluated several evidence-based small-bore feeding tube placement methods, including placement with fluoroscopy, a two-step X-ray, electromagnetic visualization, and capnography. After the evaluation, capnography was selected as the most effective method to mitigate the complications of blind insertion. In this article, the authors describe a quality improvement project involving the implementation of capnography-guided small-bore feeding tube placement to reduce complications and the incidence of lung perforation. Since the completion of the project, which took place from December 13, 2021, through April 18, 2022, no lung injuries or perforations have been reported. Capnography is a relatively simple, noninvasive, and cost-effective technology that provides nurses with a means to safely and effectively insert small-bore feeding tubes, decrease the incidence of adverse events, and improve patient care.


Subject(s)
Lung Injury , Humans , Lung Injury/prevention & control , Lung Injury/etiology , Enteral Nutrition/instrumentation , Enteral Nutrition/methods , Enteral Nutrition/nursing , Capnography , Intubation, Gastrointestinal/adverse effects , Intubation, Gastrointestinal/methods , Intubation, Gastrointestinal/nursing , Quality Improvement , Patient Safety , Trauma Centers
12.
Curr Pharm Des ; 30(27): 2179-2186, 2024.
Article in English | MEDLINE | ID: mdl-38874044

ABSTRACT

INTRODUCTION: It has been hypothesized that piperine, the main alkaloid component of black pepper, possesses a unique radioprotective effect. This study aimed to investigate the protective effect of piperine against Radiation-Induced Lung Injury (RILI) in mice. METHODS: Firstly, eighty male mice were divided into eight groups; the control group did not receive any dosage of piperine and radiation (6 Gy), and the other groups received piperine alone at doses 10, 25, and 50 mg/kg, radiation, and radiation-piperine combination (6 Gy + 10, 25, and 50 mg/kg). Animals received piperine by gavage for 7 consecutive days. To investigate the effect of piperine pretreatment in mice that were exposed to radiation, histopathological and biochemical evaluations (markers of oxidative stress) were performed. Irradiation led to an increase in oxidative stress (increase in MDA and PC). Pretreatment of piperine in all three doses in irradiated mice was able to reduce oxidative stress compared to mice that were only exposed to radiation. RESULTS: Piperine at a dose of 25 mg/kg exhibited the highest protective effect as compared to other doses. Also, in the histopathological examination, it was seen that pretreatment with piperine was able to improve the infiltration of inflammatory cells and reduce the thickness of the alveolar septum and air sac damage. CONCLUSION: The outcomes completely proved significant lung protection by piperine in mice through reducing oxidative stress. This natural compound could be considered a protective agent against lung injury induced by ionizing radiation.


Subject(s)
Alkaloids , Benzodioxoles , Oxidative Stress , Piperidines , Polyunsaturated Alkamides , Radiation-Protective Agents , Animals , Polyunsaturated Alkamides/pharmacology , Alkaloids/pharmacology , Benzodioxoles/pharmacology , Piperidines/pharmacology , Male , Mice , Radiation-Protective Agents/pharmacology , Oxidative Stress/drug effects , Lung Injury/prevention & control , Lung Injury/pathology , Lung Injury/drug therapy , Lung Injury/etiology , Dose-Response Relationship, Drug , Radiation Injuries, Experimental/prevention & control , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/drug therapy
13.
Int Immunopharmacol ; 134: 112165, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38692017

ABSTRACT

Particulate matter (PM) is considered the fundamental component of atmospheric pollutants and is associated with the pathogenesis of many respiratory diseases. Fibroblast growth factor 10 (FGF10) mediates mesenchymal-epithelial signaling and has been linked with the repair process of PM-induced lung injury (PMLI). However, the pathogenic mechanism of PMLI and the specific FGF10 protective mechanism against this injury are still undetermined. PM was administered in vivo into murine airways or in vitro to human bronchial epithelial cells (HBECs), and the inflammatory response and ferroptosis-related proteins SLC7A11 and GPX4 were assessed. The present research investigates the FGF10-mediated regulation of ferroptosis in PMLI mice models in vivo and HBECs in vitro. The results showed that FGF10 pretreatment reduced PM-mediated oxidative damage and ferroptosis in vivo and in vitro. Furthermore, FGF10 pretreatment led to reduced oxidative stress, decreased secretion of inflammatory mediators, and activation of the Nrf2-dependent antioxidant signaling. Additionally, silencing of Nrf2 using siRNA in the context of FGF10 treatment attenuated the effect on ferroptosis. Altogether, both in vivo and in vitro assessments confirmed that FGF10 protects against PMLI by inhibiting ferroptosis via the Nrf2 signaling. Thus, FGF10 can be used as a novel ferroptosis suppressor and a potential treatment target in PMLI.


Subject(s)
Ferroptosis , Fibroblast Growth Factor 10 , Lung Injury , Mice, Inbred C57BL , NF-E2-Related Factor 2 , Oxidative Stress , Particulate Matter , Signal Transduction , Ferroptosis/drug effects , NF-E2-Related Factor 2/metabolism , Animals , Particulate Matter/toxicity , Humans , Signal Transduction/drug effects , Fibroblast Growth Factor 10/metabolism , Fibroblast Growth Factor 10/genetics , Mice , Oxidative Stress/drug effects , Lung Injury/chemically induced , Lung Injury/metabolism , Lung Injury/pathology , Lung Injury/prevention & control , Male , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Cell Line , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Disease Models, Animal , Amino Acid Transport System y+
14.
BMC Pulm Med ; 24(1): 237, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38745191

ABSTRACT

BACKGROUND: Diabetes mellitus (DM) can aggravate lung ischemia-reperfusion (I/R) injury and is a significant risk factor for recipient mortality after lung transplantation. Metformin protects against I/R injury in a variety of organs. However, the effect of metformin on diabetic lung I/R injury remains unclear. Therefore, this study aimed to observe the effect and mechanism of metformin on lung I/R injury following lung transplantation in type 2 diabetic rats. METHODS: Sprague-Dawley rats were randomly divided into the following six groups: the control + sham group (CS group), the control + I/R group (CIR group), the DM + sham group (DS group), the DM + I/R group (DIR group), the DM + I/R + metformin group (DIRM group) and the DM + I/R + metformin + Compound C group (DIRMC group). Control and diabetic rats underwent the sham operation or left lung transplantation operation. Lung function, alveolar capillary permeability, inflammatory response, oxidative stress, necroptosis and the p-AMPK/AMPK ratio were determined after 24 h of reperfusion. RESULTS: Compared with the CIR group, the DIR group exhibited decreased lung function, increased alveolar capillary permeability, inflammatory responses, oxidative stress and necroptosis, but decreased the p-AMPK/AMPK ratio. Metformin improved the function of lung grafts, decreased alveolar capillary permeability, inflammatory responses, oxidative stress and necroptosis, and increased the p-AMPK/AMPK ratio. In contrast, the protective effects of metformin were abrogated by Compound C. CONCLUSIONS: Metformin attenuates lung I/R injury and necroptosis through AMPK pathway in type 2 diabetic lung transplant recipient rats.


Subject(s)
AMP-Activated Protein Kinases , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Lung Transplantation , Metformin , Necroptosis , Reperfusion Injury , Animals , Rats , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Hypoglycemic Agents/pharmacology , Lung/pathology , Lung/drug effects , Lung/metabolism , Lung Injury/prevention & control , Lung Injury/etiology , Lung Injury/metabolism , Metformin/pharmacology , Necroptosis/drug effects , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control , Signal Transduction/drug effects
15.
Biomed Pharmacother ; 175: 116674, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703509

ABSTRACT

Numerous cases of lung injury caused by viral infection were reported during the coronavirus disease-19 pandemic. While there have been significant efforts to develop drugs that block viral infection and spread, the development of drugs to reduce or reverse lung injury has been a lower priority. This study aimed to identify compounds from a library of compounds that prevent viral infection that could reduce and prevent lung epithelial cell damage. We investigated the cytotoxicity of the compounds, their activity in inhibiting viral spike protein binding to cells, and their activity in reducing IL-8 production in lung epithelial cells damaged by amodiaquine (AQ). We identified N-(4-(4-methoxyphenoxy)-3-methylphenyl)-N-methylacetamide (MPoMA) as a non-cytotoxic inhibitor against viral infection and AQ-induced cell damage. MPoMA inhibited the expression of IL-8, IL-6, IL-1ß, and fibronectin induced by AQ and protected against AQ-induced morphological changes. However, MPoMA did not affect basal IL-8 expression in lung epithelial cells in the absence of AQ. Further mechanistic analysis confirmed that MPoMA selectively promoted the proteasomal degradation of inflammatory mediator p65, thereby reducing intracellular p65 expression and p65-mediated inflammatory responses. MPoMA exerted potent anti-inflammatory and protective functions in epithelial cells against LPS-induced acute lung injury in vivo. These findings suggest that MPoMA may have beneficial effects in suppressing viral infection and preventing lung epithelial cell damage through the degradation of p65 and inhibition of the production of inflammatory cytokines.


Subject(s)
Epithelial Cells , Animals , Humans , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Mice , Lung/pathology , Lung/drug effects , Lung/metabolism , Transcription Factor RelA/metabolism , COVID-19 Drug Treatment , A549 Cells , SARS-CoV-2/drug effects , COVID-19/prevention & control , Proteolysis/drug effects , Lung Injury/prevention & control , Lung Injury/pathology , Lung Injury/metabolism , Lung Injury/virology , Male , Acute Lung Injury/prevention & control , Acute Lung Injury/pathology , Acute Lung Injury/metabolism , Acetamides/pharmacology
16.
Surgery ; 176(2): 499-510, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38811326

ABSTRACT

BACKGROUND: Emodin, a natural anthraquinone derivative found in various Chinese medicinal herbs, has been proved to be an effective therapeutic agent in the treatment of many diseases. However, its effect on lung injury after intestinal ischemia/reperfusion injury remains unknown. This research was designed to investigate whether emodin protects against intestinal ischemia/reperfusion-induced lung injury and to elucidate the underlying molecular mechanisms in vivo and in vitro. METHODS: Intestinal ischemia/reperfusion injury was induced by occluding the superior mesenteric artery in mice, and mouse lung epithelial-12 cells were subjected to oxygen-glucose deprivation and reoxygenation to establish an in vitro model. RESULTS: Our data indicated that emodin treatment reduced intestinal ischemia/reperfusion-induced oxidative stress, inflammation and apoptosis in lung tissues and alleviated lung injury. However, the protective effects of emodin on intestinal ischemia/reperfusion-induced lung injury were reversed by the protein kinase B inhibitor triciribine or the heme oxygenase-1 inhibitor tin protoporphyrin IX. The protein kinase inhibitor triciribine also downregulated the expression of heme oxygenase-1. CONCLUSION: In conclusion, our data suggest that emodin treatment protects against intestinal ischemia/reperfusion-induced lung injury by enhancing heme oxygenase-1 expression via activation of the PI3K/protein kinase pathway. Emodin may act as a potential therapeutic agent for the prevention and treatment of lung injury induced by intestinal ischemia/reperfusion.


Subject(s)
Emodin , Heme Oxygenase-1 , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Reperfusion Injury , Signal Transduction , Up-Regulation , Animals , Emodin/pharmacology , Emodin/therapeutic use , Reperfusion Injury/prevention & control , Reperfusion Injury/metabolism , Reperfusion Injury/etiology , Reperfusion Injury/drug therapy , Mice , Proto-Oncogene Proteins c-akt/metabolism , Heme Oxygenase-1/metabolism , Male , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , Intestines/blood supply , Intestines/pathology , Intestines/drug effects , Mice, Inbred C57BL , Lung Injury/etiology , Lung Injury/prevention & control , Lung Injury/metabolism , Lung Injury/drug therapy , Lung Injury/pathology , Disease Models, Animal , Oxidative Stress/drug effects , Membrane Proteins
18.
Br J Pharmacol ; 181(17): 3215-3231, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38741475

ABSTRACT

BACKGROUND AND PURPOSE: Nicorandil, a selective opener of potassium channels, used to treat angina, has drawn attention for its potential in mitigating lung injury, positioning it as a promising therapeutic approach to treat drug-induced lung toxicity. This study aimed to explore the protective role of nicorandil in arsenic trioxide (ATO)-induced lung injury and to elucidate the underlying mechanistic pathways. EXPERIMENTAL APPROACH: We assessed the effects of nicorandil (15 mg·kg-1, p.o.) in a rat model of pulmonary injury induced by ATO (5 mg·kg-1, i.p.). The assessment included oxidative stress biomarkers, inflammatory cytokine levels, and other biomarkers, including sirtuin-1, sirtuin-3, STAT3, TFAM, and JAK in lung tissue. Histological examination using H&E staining and molecular investigations using western blotting and PCR techniques were conducted. KEY RESULTS: In our model of lung injury, treatment with nicorandil ameliorated pathological changes as seen with H&E staining, reduced tissue levels of toxicity markers, and exerted significant antioxidant and anti-inflammatory actions. On a molecular level, treatment with nicorandil down-regulated JAK, STAT3, PPARγ, Nrf2, VEGF, p53, and micro-RNA 132 while up-regulating Sirt1, 3, TFAM, AMPK, and ERR-α in lung tissue. CONCLUSIONS AND IMPLICATIONS: The results presented here show nicorandil as a significant agent in attenuating lung injury induced by ATO in a rodent model. Nonetheless, further clinical studies are warranted to strengthen these findings.


Subject(s)
Arsenic Trioxide , Janus Kinase 1 , Lung Injury , MicroRNAs , Nicorandil , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , STAT3 Transcription Factor , Signal Transduction , Sirtuin 1 , Animals , Nicorandil/pharmacology , STAT3 Transcription Factor/metabolism , Sirtuin 1/metabolism , Sirtuin 1/genetics , Male , Signal Transduction/drug effects , Rats , Arsenic Trioxide/pharmacology , Arsenic Trioxide/toxicity , MicroRNAs/metabolism , MicroRNAs/genetics , Janus Kinase 1/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Lung Injury/chemically induced , Lung Injury/metabolism , Lung Injury/drug therapy , Lung Injury/pathology , Lung Injury/prevention & control , Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Rats, Wistar , Rats, Sprague-Dawley , Oxidative Stress/drug effects
20.
Nature ; 628(8009): 835-843, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38600381

ABSTRACT

Severe influenza A virus (IAV) infections can result in hyper-inflammation, lung injury and acute respiratory distress syndrome1-5 (ARDS), for which there are no effective pharmacological therapies. Necroptosis is an attractive entry point for therapeutic intervention in ARDS and related inflammatory conditions because it drives pathogenic lung inflammation and lethality during severe IAV infection6-8 and can potentially be targeted by receptor interacting protein kinase 3 (RIPK3) inhibitors. Here we show that a newly developed RIPK3 inhibitor, UH15-38, potently and selectively blocked IAV-triggered necroptosis in alveolar epithelial cells in vivo. UH15-38 ameliorated lung inflammation and prevented mortality following infection with laboratory-adapted and pandemic strains of IAV, without compromising antiviral adaptive immune responses or impeding viral clearance. UH15-38 displayed robust therapeutic efficacy even when administered late in the course of infection, suggesting that RIPK3 blockade may provide clinical benefit in patients with IAV-driven ARDS and other hyper-inflammatory pathologies.


Subject(s)
Lung Injury , Necroptosis , Orthomyxoviridae Infections , Protein Kinase Inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases , Animals , Female , Humans , Male , Mice , Alveolar Epithelial Cells/pathology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/virology , Alveolar Epithelial Cells/metabolism , Influenza A virus/classification , Influenza A virus/drug effects , Influenza A virus/immunology , Influenza A virus/pathogenicity , Lung Injury/complications , Lung Injury/pathology , Lung Injury/prevention & control , Lung Injury/virology , Mice, Inbred C57BL , Necroptosis/drug effects , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/prevention & control , Respiratory Distress Syndrome/virology
SELECTION OF CITATIONS
SEARCH DETAIL