Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 852
Filtrer
1.
Cell Biol Toxicol ; 40(1): 70, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39136896

RÉSUMÉ

Paraquat poisoning results in significant pulmonary damage, but current treatments are only minimally effective in repairing the injured lung tissues. Recent research has highlighted the promise of using stem cell therapy, namely mesenchymal stem cells, as a new method for treating paraquat toxicity. These cells have shown effectiveness in decreasing inflammation, apoptosis, and fibrosis in the mice lungs subjected to paraquat. The therapeutic implications of mesenchymal stem cells are believed to arise from their release of bioactive proteins and their capacity to regulate inflammatory responses. However, additional clinical study is required to validate these therapies' efficacy. This review thoroughly explores the pathophysiology of paraquat poisoning and the properties of mesenchymal stem cells. Additionally, it critically assesses the long-term safety and effectiveness of mesenchymal stem cell therapies, which is crucial for developing more dependable and effective treatment protocols. In summary, although mesenchymal stem cells offer promising prospects for treating lung injuries, more investigations are required to optimize their therapeutic promise and ensure their safe clinical application in the context of paraquat poisoning.


Sujet(s)
Lésion pulmonaire , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Paraquat , Paraquat/toxicité , Animaux , Humains , Transplantation de cellules souches mésenchymateuses/méthodes , Lésion pulmonaire/thérapie , Lésion pulmonaire/induit chimiquement , Poumon/anatomopathologie , Souris
2.
Int J Immunopathol Pharmacol ; 38: 3946320241272642, 2024.
Article de Anglais | MEDLINE | ID: mdl-39096175

RÉSUMÉ

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.


Sujet(s)
Flavones , Inflammasomes , Lésion pulmonaire , Nicotine , Animaux , Mâle , Rats , Flavones/pharmacologie , Protéine O1 à motif en tête de fourche , Rayons gamma , Inflammasomes/métabolisme , Inflammasomes/effets des médicaments et des substances chimiques , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Poumon/anatomopathologie , Poumon/effets des radiations , Lésion pulmonaire/métabolisme , Lésion pulmonaire/prévention et contrôle , Facteur-2 apparenté à NF-E2/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Nicotine/pharmacologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinases/métabolisme , Rat Wistar , Transduction du signal/effets des médicaments et des substances chimiques
3.
Immun Inflamm Dis ; 12(8): e1343, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39092750

RÉSUMÉ

The involvement of neutrophils in the lungs during the recovery phase of coronavirus disease 2019 (COVID-19) is not well defined mainly due to the limited accessibility of lung tissues from COVID-19 survivors. The lack of an appropriate small animal model has affected the development of effective therapeutic strategies. We here developed a long COVID mouse model to study changes in neutrophil phenotype and association with lung injury. Our data shows persistent neutrophil recruitment and neutrophil extracellular trap formation in the lungs for up to 30 days post-infection which correlates with lung fibrosis and inflammation.


Sujet(s)
COVID-19 , Modèles animaux de maladie humaine , Pièges extracellulaires , Poumon , Granulocytes neutrophiles , SARS-CoV-2 , Animaux , Pièges extracellulaires/immunologie , COVID-19/immunologie , COVID-19/complications , Souris , Granulocytes neutrophiles/immunologie , SARS-CoV-2/immunologie , SARS-CoV-2/physiologie , Poumon/anatomopathologie , Poumon/immunologie , Poumon/virologie , Lésion pulmonaire/immunologie , Lésion pulmonaire/virologie , Lésion pulmonaire/anatomopathologie , Lésion pulmonaire/étiologie , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , Infiltration par les neutrophiles/immunologie , Humains , Fibrose pulmonaire/immunologie , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/étiologie
4.
J Biochem Mol Toxicol ; 38(8): e23790, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39108137

RÉSUMÉ

Pulmonary injury is one of the key restricting factors for the therapy of malignancies with chemotherapy or following radiotherapy for chest cancers. The lung is a sensitive organ to some severely toxic antitumor drugs, consisting of bleomycin and alkylating agents. Furthermore, treatment with radiotherapy may drive acute and late adverse impacts on the lung. The major consequences of radiotherapy and chemotherapy in the lung are pneumonitis and fibrosis. Pneumonitis may arise some months to a few years behind cancer therapy. However, fibrosis is a long-term effect that appears years after chemo/or radiotherapy. Several mechanisms such as oxidative stress and severe immune reactions are implicated in the progression of pulmonary fibrosis. Epithelial-mesenchymal transition (EMT) is offered as a pivotal mechanism for lung fibrosis behind chemotherapy and radiotherapy. It seems that pulmonary fibrosis is the main consequence of EMT after chemo/radiotherapy. Several biological processes, consisting of the liberation of pro-inflammatory and pro-fibrosis molecules, oxidative stress, upregulation of nuclear factor of κB and Akt, epigenetic changes, and some others, may participate in EMT and pulmonary fibrosis behind cancer therapy. In this review, we aim to discuss how chemotherapy or radiotherapy may promote EMT and lung fibrosis. Furthermore, we review potential targets and effective agents to suppress EMT and lung fibrosis after cancer therapy.


Sujet(s)
Chimioradiothérapie , Transition épithélio-mésenchymateuse , Fibrose pulmonaire , Humains , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/étiologie , Chimioradiothérapie/effets indésirables , Animaux , Stress oxydatif/effets des médicaments et des substances chimiques , Lésion pulmonaire/étiologie , Lésion pulmonaire/anatomopathologie , Lésion pulmonaire/induit chimiquement , Lésion pulmonaire/métabolisme
5.
Mol Biol Rep ; 51(1): 891, 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39110355

RÉSUMÉ

BACKGROUND: Peptide transporter 1 (PepT1) transports bacterial oligopeptide products and induces inflammation of the bowel. Nutritional peptides compete for the binding of intestinal bacterial products to PepT1. We investigated the mechanism of short-peptide-based enteral nutrition (SPEN) on the damage to the gut caused by the bacterial oligopeptide product muramyl dipeptide (MDP), which is transported by PepT1. The gut-lung axis is a shared mucosal immune system, and immune responses and disorders can affect the gut-respiratory relationship. METHODS AND RESULTS: Sprague-Dawley rats were gavaged with solutions containing MDP, MDP + SPEN, MDP + intact-protein-based enteral nutrition (IPEN), glucose as a control, or glucose with GSK669 (a NOD2 antagonist). Inflammation, mitochondrial damage, autophagy, and apoptosis were explored to determine the role of the PepT1-nucleotide-binding oligomerization domain-containing protein 2 (NOD2)-beclin-1 signaling pathway in the small intestinal mucosa. MDP and proinflammatory factors of lung tissue were explored to determine that MDP can migrate to lung tissue and cause inflammation. Induction of proinflammatory cell accumulation and intestinal damage in MDP gavage rats was associated with increased NOD2 and Beclin-1 mRNA expression. IL-6 and TNF-α expression and apoptosis were increased, and mitochondrial damage was severe, as indicated by increased mtDNA in the MDP group compared with controls. MDP levels and expression of proinflammatory factors in lung tissue increased in the MDP group compared with the control group. SPEN, but not IPEN, eliminated these impacts. CONCLUSIONS: Gavage of MDP to rats resulted in damage to the gut-lung axis. SPEN reverses the adverse effects of MDP. The PepT1-NOD2-beclin-1 pathway plays a role in small intestinal inflammation, mitochondrial damage, autophagy, and apoptosis.


Sujet(s)
Acétylmuramyl alanyl isoglutamine , Bécline-1 , Nutrition entérale , Lésion pulmonaire , Protéine adaptatrice de signalisation NOD2 , Transporteur-1 de peptides , Rat Sprague-Dawley , Transduction du signal , Animaux , Transporteur-1 de peptides/métabolisme , Transporteur-1 de peptides/génétique , Rats , Bécline-1/métabolisme , Bécline-1/génétique , Protéine adaptatrice de signalisation NOD2/métabolisme , Protéine adaptatrice de signalisation NOD2/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Lésion pulmonaire/métabolisme , Mâle , Acétylmuramyl alanyl isoglutamine/pharmacologie , Nutrition entérale/méthodes , Apoptose/effets des médicaments et des substances chimiques , Muqueuse intestinale/métabolisme , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/anatomopathologie , Autophagie/effets des médicaments et des substances chimiques , Poumon/métabolisme , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Inflammation/métabolisme
6.
Respir Res ; 25(1): 299, 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39113018

RÉSUMÉ

BACKGROUND: Although recent studies provide mechanistic understanding to the pathogenesis of radiation induced lung injury (RILI), rare therapeutics show definitive promise for treating this disease. Type II alveolar epithelial cells (AECII) injury in various manner results in an inflammation response to initiate RILI. RESULTS: Here, we reported that radiation (IR) up-regulated the TNKS1BP1, causing progressive accumulation of the cellular senescence by up-regulating EEF2 in AECII and lung tissue of RILI mice. Senescent AECII induced Senescence-Associated Secretory Phenotype (SASP), consequently activating fibroblasts and macrophages to promote RILI development. In response to IR, elevated TNKS1BP1 interacted with and decreased CNOT4 to suppress EEF2 degradation. Ectopic expression of EEF2 accelerated AECII senescence. Using a model system of TNKS1BP1 knockout (KO) mice, we demonstrated that TNKS1BP1 KO prevents IR-induced lung tissue senescence and RILI. CONCLUSIONS: Notably, this study suggested that a regulatory mechanism of the TNKS1BP1/CNOT4/EEF2 axis in AECII senescence may be a potential strategy for RILI.


Sujet(s)
Pneumocytes , Vieillissement de la cellule , Souris de lignée C57BL , Souris knockout , Animaux , Souris , Vieillissement de la cellule/effets des radiations , Vieillissement de la cellule/physiologie , Pneumocytes/métabolisme , Pneumocytes/effets des radiations , Pneumocytes/anatomopathologie , Lésion pulmonaire/métabolisme , Lésion pulmonaire/génétique , Lésion pulmonaire/anatomopathologie , Elongation Factor 2 Kinase/métabolisme , Elongation Factor 2 Kinase/génétique , Humains , Lésions radiques expérimentales/métabolisme , Lésions radiques expérimentales/anatomopathologie , Lésions radiques expérimentales/génétique , Cellules cultivées , Mâle
7.
Sci Immunol ; 9(98): eado1227, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39093958

RÉSUMÉ

The lung is constantly exposed to airborne pathogens and particles that can cause alveolar damage. Hence, appropriate repair responses are essential for gas exchange and life. Here, we deciphered the spatiotemporal trajectory and function of an atypical population of macrophages after lung injury. Post-influenza A virus (IAV) infection, short-lived monocyte-derived Ly6G-expressing macrophages (Ly6G+ Macs) were recruited to the alveoli of lung perilesional areas. Ly6G+ Macs engulfed immune cells, exhibited a high metabolic potential, and clustered with alveolar type 2 epithelial cells (AT2s) in zones of active epithelial regeneration. Ly6G+ Macs were partially dependent on granulocyte-macrophage colony-stimulating factor and interleukin-4 receptor signaling and were essential for AT2-dependent alveolar regeneration. Similar macrophages were recruited in other models of injury and in the airspaces of lungs from patients with suspected pneumonia. This study identifies perilesional alveolar Ly6G+ Macs as a spatially restricted, short-lived macrophage subset promoting epithelial regeneration postinjury, thus representing an attractive therapeutic target for treating lung damage.


Sujet(s)
Antigènes Ly , Lésion pulmonaire , Macrophages alvéolaires , Souris de lignée C57BL , Régénération , Animaux , Antigènes Ly/métabolisme , Antigènes Ly/immunologie , Souris , Régénération/immunologie , Lésion pulmonaire/immunologie , Macrophages alvéolaires/immunologie , Mâle , Humains , Femelle , Infections à Orthomyxoviridae/immunologie , Alvéoles pulmonaires/immunologie , Virus de la grippe A/immunologie , Virus de la grippe A/physiologie
8.
Exerc Immunol Rev ; 30: 49-62, 2024.
Article de Anglais | MEDLINE | ID: mdl-39094180

RÉSUMÉ

Moderate exercise is effective for maintaining or improving overall health. However, excessive exercise that exhausts the adaptive reserve of the body or its ability to positively respond to training stimuli can induce tissue damage and dysfunction of multiple organs and systems. Tissue injury, inflammation, and oxidative stress are reportedly induced in the skeletal muscles, liver, and kidneys after exercise. However, the precise mechanisms underlying acute tissue injury after intense exercise have not yet been fully elucidated. Studies using various experimental models of acute tissue injury, other than intense exercise, have demonstrated infiltration of inflammatory cells, including neutrophils and macrophages. These cells infiltrate injured tissues and induce inflammatory and oxidative stress responses by producing inflammatory cytokines and reactive oxygen species, thereby exacerbating tissue injury. In addition to the activation of blood neutrophils and increase in their levels during and/or after prolonged or intense exercise, chemokines that contribute to leukocyte migration are released, facilitating the migration of neutrophils and monocytes into tissues. Therefore, neutrophils and macrophages, activated by exhaustive exercise, may infiltrate tissues and contribute to exhaustive exercise-induced tissue injury. Recently, the contributions of neutrophils and macrophages to various tissue injuries caused by exhaustive exercise have been reported. In this review, we summarize the involvement of neutrophils and monocytes/macrophages in exhaustive exercise-induced non-skeletal muscle tissue injury. In addition, we present novel data demonstrating the contribution of neutrophils and macrophages to exhaustive exercise-induced cardiac and pulmonary injuries. Our study findings and the evidence presented in this review suggest that neutrophils and macrophages may play pivotal roles in exhaustive exercise-induced tissue injuries.


Sujet(s)
Exercice physique , Macrophages , Granulocytes neutrophiles , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Humains , Macrophages/immunologie , Exercice physique/physiologie , Animaux , Foie/immunologie , Foie/anatomopathologie , Foie/métabolisme , Foie/traumatismes , Lésion pulmonaire/immunologie , Lésion pulmonaire/étiologie , Lésions traumatiques du coeur/immunologie , Lésions traumatiques du coeur/étiologie , Stress oxydatif , Rein/immunologie , Rein/anatomopathologie
9.
Allergol Immunopathol (Madr) ; 52(4): 53-59, 2024.
Article de Anglais | MEDLINE | ID: mdl-38970265

RÉSUMÉ

BACKGROUND: Pulmonary fibrosis is a pathological hallmark of lung injury. It is an aggressive disease that replaces normal lung parenchyma by fibrotic tissue. The transforming growth factor-beta-mothers against decapentaplegic homolog 3 (TGF-ß1-Smad3) signaling pathway plays a key role in regulating lung fibrosis. Decorin (DCN), a small leucine-rich proteoglycan, has a modulatory effect on the immune system by reversibly binding with TGF-ß and reducing its bioavailability. Mesenchymal stem cell (MSC) therapy is a new strategy that has an immune-modulatory capacity. OBJECTIVE: The aim of this study was to introduce a new therapeutic approach to harness remodeling in injured lung. MATERIAL AND METHODS: Bone marrow MSCs were isolated and transduced by decorin gene. Lung injury was induced by bleomycin and mice were treated with MSCs, MSCs-decorin, and decorin. Then, oxidative stress biomarkers, remodeling biomarkers, bronchoalveolar lavage cells, and histopathology study were conducted. RESULTS: Reduced catalase and superoxide dismutase increased due to treatments. Elevated malondialdehyde, hydroxyproline, TGF-ß levels, and polymorphonuclear cells count decreased in the treated groups. Additionally, the histopathology of lung tissues showed controlled inflammation and fibrosis. CONCLUSION: Transfected decorin gene to MSCs and used cell therapy could control remodeling and bleomycin-induced lung injury.


Sujet(s)
Bléomycine , Décorine , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Fibrose pulmonaire , Décorine/génétique , Décorine/métabolisme , Animaux , Souris , Fibrose pulmonaire/immunologie , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/thérapie , Lésion pulmonaire/induit chimiquement , Lésion pulmonaire/thérapie , Lésion pulmonaire/immunologie , Lésion pulmonaire/génétique , Transduction génétique , Stress oxydatif , Cellules cultivées , Modèles animaux de maladie humaine , Mâle , Humains
10.
Sci Rep ; 14(1): 15437, 2024 07 04.
Article de Anglais | MEDLINE | ID: mdl-38965293

RÉSUMÉ

We aimed to determine the clinical characteristics of patient self-inflicted lung injury (P-SILI)-associated pneumothorax/pneumomediastinum, to reveal its risk factors, and to assess its impact on severe COVID-19 cases. In total, 229 patients were included in this case-control study. They were randomly divided into either the case group or the control group as per the inclusion and exclusion criteria. The two groups were further analyzed to reveal the risk factors of spontaneous pneumothorax/pneumomediastinum (SP/P). Finally, risk factors for death were analyzed in the case group and the relationship between death and SP/P was also analyzed among all patients. The mean age of patients was 59.69 ± 17.01 years, most of them were male (74.2%), and 62.0% of them had comorbidities upon admission. A respiratory rate higher than 30 BPM was a risk factor for SP/P (OR 7.186, 95% CI 2.414-21.391, P < 0.001). Patients with delayed intubation due to early application of HFNC or NIV had a higher mortality rate when they developed SP/P (P < 0.05). Additionally, advanced age increased the risk of death (P < 0.05). Finally, SP/P may be a risk factor for death among patients with severe COVID-19 (OR 2.047). P-SILI occurs in severe COVID-19 with acute respiratory failure. It is necessary to identify the risk factors of P-SILI, the indicators of severe P-SILI, and the preventive measures.


Sujet(s)
COVID-19 , Emphysème médiastinal , Pneumothorax , Humains , COVID-19/complications , COVID-19/mortalité , Mâle , Adulte d'âge moyen , Femelle , Études cas-témoins , Facteurs de risque , Emphysème médiastinal/étiologie , Pneumothorax/étiologie , Sujet âgé , Adulte , Lésion pulmonaire/étiologie , Comportement auto-agressif/complications , SARS-CoV-2
11.
J Cardiothorac Surg ; 19(1): 445, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39004745

RÉSUMÉ

BACKGROUND: Penetrating thoracic injuries have a significant risk of morbi-mortality. Despite the advancements in damage control methods, a subset of patients with severe pulmonary vascular lesions and bronchial injuries persists. In some of these cases, post-traumatic pneumonectomy is required, and perioperative extracorporeal membrane oxygenation (ECMO) support may be required due to right ventricular failure and respiratory failure. CASE DESCRIPTION: A male was brought to the emergency department (ED) with a penetrating thoracic injury, presenting with massive right hemothorax and active bleeding that required ligation of the right pulmonary hilum to control the bleeding. Subsequently, he developed right ventricular dysfunction and ARDS, necessitating a dynamic hybrid ECMO configuration to support his condition and facilitate recovery. CONCLUSIONS: Penetrating thoracic injuries with severe pulmonary vascular lesions may need pneumonectomy to control bleeding. ECMO support reduces the associated mortality by decreasing the complications rate. A multidisciplinary team is essential to achieve good outcomes in severe compromised patients.


Sujet(s)
Oxygénation extracorporelle sur oxygénateur à membrane , Pneumonectomie , Humains , Oxygénation extracorporelle sur oxygénateur à membrane/méthodes , Mâle , Lésion pulmonaire/chirurgie , Lésion pulmonaire/étiologie , Adulte , Blessures du thorax/chirurgie , Blessures du thorax/complications , Plaies pénétrantes/chirurgie , Hémothorax/étiologie , Hémothorax/chirurgie , Soins postopératoires/méthodes
12.
Ecotoxicol Environ Saf ; 282: 116711, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39002383

RÉSUMÉ

Silicosis, caused by silica exposure, is the most widespread and deadliest occupational disease. However, effective treatments are lacking. Therefore, it is crucial to elucidate the mechanisms and targets involved in the development of silicosis. We investigated the basic processes of silicosis development and onset at different exposure durations (2 or 4 weeks) using various techniques such as histopathology, immunohistochemistry, Enzyme linked immunosorbent assay(ELISA),16 S rRNA, and untargeted metabolomics.These results indicate that exposure to silica leads to progressive damage to lung tissue with significant deterioration observed over time. Time-dependent cytokines such as the IL-4, IL-13, and IL-6 are detected in lung lavage fluid, the model group consistently exhibited elevated levels of these cytokines, indicating a persistent and worsening inflammatory response in the lungs. Meanwhile, HE and Masson results show that 4-week exposure to silica causes more obvious lung injury and pulmonary fibrosis. Besides, the model group consistently exhibited a distinct lung bacterial population, known as the Lachnospiraceae_NK4A136_group, regardless of exposure duration. However, with increasing exposure duration, specific temporal changes were observed in lung bacterial populations, including Haliangium, Allobaculum, and Sandaracinus (at 4 weeks; p < 0.05). Furthermore, our study revealed a strong correlation between the mechanism of silica-induced lung injury and three factors: oxidative stress, impaired lipid metabolism, and imbalanced amino acid metabolism. We observed a close correlation between cytokine levels, changes in lung microbiota, and metabolic disturbances during various exposure periods. These findings propose that a possible mechanism of silica-induced lung injury involves the interplay of cytokines, lung microbiota, and metabolites.


Sujet(s)
Cytokines , Lésion pulmonaire , Poumon , Microbiote , Silice , Silice/toxicité , Animaux , Poumon/microbiologie , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Microbiote/effets des médicaments et des substances chimiques , Lésion pulmonaire/induit chimiquement , Lésion pulmonaire/microbiologie , Lésion pulmonaire/anatomopathologie , Cytokines/métabolisme , Mâle , Silicose/métabolisme , Liquide de lavage bronchoalvéolaire/composition chimique
13.
J Hazard Mater ; 476: 135103, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-38972203

RÉSUMÉ

An earlier study found that respiratory cadmium chloride (CdCl2) exposure caused COPD-like lung injury. This study aimed to explore whether mitochondrial dysfunction-mediated alveolar epithelial senescence is involved in CdCl2-induced COPD-like lung injury. Adult C57BL/6 mice were exposed to CdCl2 (10 mg/L) aerosol for six months. Beta-galactosidase-positive cells, p21 and p16 were increased in CdCl2-exposed mouse lungs. The in vitro experiments showed that γ-H2AX was elevated in CdCl2-exposed alveolar epithelial cells. The cGAS-STING pathway was activated in CdCl2-exposed alveolar epithelial cells and mouse lungs. Cxcl1, Cxcl9, Il-10, Il-1ß and Mmp2, several senescence-associated secretory phenotypes (SASP), were upregulated in CdCl2-exposed alveolar epithelial cells. Mechanistically, CdCl2 exposure caused SIRT3 reduction and mitochondrial dysfunction in mouse lungs and alveolar epithelial cells. The in vitro experiment found that Sirt3 overexpression attenuated CdCl2-induced alveolar epithelial senescence and SASP. The in vivo experiments showed that Sirt3 gene knockout exacerbated CdCl2-induced alveolar epithelial senescence, alveolar structure damage, airway inflammation and pulmonary function decline. NMN, an NAD+ precursor, attenuated CdCl2-induced alveolar epithelial senescence and SASP in mouse lungs. Moreover, NMN supplementation prevented CdCl2-induced COPD-like alveolar structure damage, epithelial-mesenchymal transition and pulmonary function decline. These results suggest that mitochondrial dysfunction-associated alveolar epithelial senescence is involved in CdCl2-induced COPD-like lung injury.


Sujet(s)
Vieillissement de la cellule , Souris de lignée C57BL , Mitochondries , Broncho-pneumopathie chronique obstructive , Animaux , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Broncho-pneumopathie chronique obstructive/anatomopathologie , Broncho-pneumopathie chronique obstructive/induit chimiquement , Broncho-pneumopathie chronique obstructive/métabolisme , Pneumocytes/effets des médicaments et des substances chimiques , Pneumocytes/métabolisme , Mâle , Lésion pulmonaire/induit chimiquement , Lésion pulmonaire/anatomopathologie , Sirtuine-3/métabolisme , Sirtuine-3/génétique , Souris , Souris knockout
14.
J Physiol Pharmacol ; 75(3)2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-39042394

RÉSUMÉ

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.


Sujet(s)
Animaux nouveau-nés , Dexmédétomidine , Hyperoxie , Lésion pulmonaire , Poumon , Protéine-3 de la famille des NLR contenant un domaine pyrine , Pyroptose , Rat Sprague-Dawley , Animaux , Dexmédétomidine/pharmacologie , Dexmédétomidine/usage thérapeutique , Hyperoxie/métabolisme , Hyperoxie/complications , Hyperoxie/traitement médicamenteux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Pyroptose/effets des médicaments et des substances chimiques , Lésion pulmonaire/métabolisme , Lésion pulmonaire/prévention et contrôle , Lésion pulmonaire/anatomopathologie , Lésion pulmonaire/traitement médicamenteux , Rats , Protéines de liaison aux phosphates/métabolisme , Protéines adaptatrices de signalisation CARD/métabolisme , Caspase-1/métabolisme , Interleukine-18/métabolisme , Agonistes des récepteurs alpha-2 adrénergiques/pharmacologie , Agonistes des récepteurs alpha-2 adrénergiques/usage thérapeutique , Mâle , Gasdermines
15.
Biochem Pharmacol ; 227: 116418, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38996928

RÉSUMÉ

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.


Sujet(s)
Protéines et peptides de signalisation intracellulaire , MAP Kinase Kinase Kinases , Souris de lignée C57BL , Facteur de transcription NF-kappa B , Sepsie , Ubiquitin-specific proteases , Animaux , Sepsie/métabolisme , MAP Kinase Kinase Kinases/métabolisme , MAP Kinase Kinase Kinases/génétique , Facteur de transcription NF-kappa B/métabolisme , Souris , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Mâle , Humains , Ubiquitin-specific proteases/métabolisme , Ubiquitin-specific proteases/génétique , Transduction du signal/physiologie , Ubiquitination , Lésion pulmonaire/métabolisme , Lésion pulmonaire/étiologie , Lésion pulmonaire/prévention et contrôle , Système de signalisation des MAP kinases/physiologie
16.
Chem Res Toxicol ; 37(8): 1415-1427, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39078936

RÉSUMÉ

The outbreak of e-cigarette or vaping use-associated lung injury (EVALI) in the United States in 2019 led to a total of 2807 hospitalizations with 68 deaths. While the exact causes of this vaping-related lung illness are still being debated, laboratory analyses of products from victims of EVALI have shown that vitamin E acetate (VEA), an additive in some tetrahydrocannabinol (THC)-containing products, is strongly linked to the EVALI outbreak. Because of its similar appearance and viscosity to pure THC oil, VEA was used as a diluent agent in cannabis oils in illicit markets. A potential mechanism for EVALI may involve VEA's thermal decomposition product, ketene, a highly poisonous gas, being generated under vaping conditions. In this study, a novel approach was developed to evaluate ketene production from VEA vaping under measurable temperature conditions in real-world devices. Ketene in generated aerosols was captured by two different chemical agents and analyzed by gas chromatography-mass spectrometry (GC-MS) and liquid chromatography with tandem mass spectrometry (LC-MS/MS). The LC-MS/MS method takes advantage of the high sensitivity and specificity of tandem mass spectrometry and appears to be more suitable than GC-MS for the analysis of large batches of samples. Our results confirmed the formation of ketene when VEA was vaped. The production of ketene increased with repeat puffs and showed a correlation to temperatures (200 to 500 °C) measured within vaping devices. Device battery power strength, which affects the heating temperature, plays an important role in ketene formation. In addition to ketene, the organic oxidant duroquinone was also obtained as another thermal degradation product of VEA. Ketene was not detected when vitamin E was vaped under the same conditions, confirming the importance of the acetate group for its generation.


Sujet(s)
Dispositifs électroniques d'administration de nicotine , Éthylènes , Cétones , Vapotage , Vapotage/effets indésirables , Cétones/composition chimique , Cétones/analyse , Éthylènes/composition chimique , Humains , Santé publique , Vitamine E/composition chimique , Vitamine E/analyse , Lésion pulmonaire/étiologie , Lésion pulmonaire/induit chimiquement , Chromatographie gazeuse-spectrométrie de masse
17.
Sci Total Environ ; 948: 174674, 2024 Oct 20.
Article de Anglais | MEDLINE | ID: mdl-39002594

RÉSUMÉ

Nanoplastics (NPs) are a common type of degraded plastic material associated with adverse health effects such as pulmonary injury. However, the molecular mechanism(s) underlying lung injury as caused by NPs remains uncertain. Thus, we herein investigated the pulmonary toxicity of NPs on RAW264.7 cells and C57BL/6 mice. Our in vitro study indicated that NPs induced oxidative stress, cell death, inflammation, and the activation of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING)-signaling pathway. Mice in our in vivo study displayed significant pulmonary fibrosis, inflammation, apoptosis, necrosis, and excessive double-stranded DNA release into serum and bronchoalveolar lavage fluid. Our mechanistic exploration uncovered cGAS-STING-signaling activation as the leading cause of NPs-induced pulmonary fibrosis. The current study opens an avenue toward elucidating the role of the cGAS-STING-signaling pathway in NPs-induced pulmonary injury.


Sujet(s)
ADN mitochondrial , Lésion pulmonaire , Nucleotidyltransferases , Transduction du signal , Animaux , Souris , Lésion pulmonaire/induit chimiquement , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Souris de lignée C57BL , Nanoparticules/toxicité , Nucleotidyltransferases/métabolisme , Nucleotidyltransferases/génétique , Cellules RAW 264.7 , Transduction du signal/effets des médicaments et des substances chimiques
18.
J Ethnopharmacol ; 334: 118584, 2024 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-39019418

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Fuzheng-Qushi decoction (FZQS) is a practical Chinese herbal formula for relieving cough and fever. Therefore, the action and specific molecular mechanism of FZQS in the treatment of lung injury with cough and fever as the main symptoms need to be further investigated. AIMS OF THE STUDY: To elucidate the protective effects of FZQS against lung injury in mice and reveal its potential targets and key biological pathways for the treatment of lung injury based on transcriptomics, microbiomics, and untargeted metabolomics analyses. MATERIALS AND METHODS: Lipopolysaccharide (LPS) was used to induce a mouse model of lung injury, followed by the administration of FZQS. ELISA was used to detect IL-1ß, IL-6, IL-17A, IL-4, IL-10, and TNF-α, in mouse lung tissues. Macrophage polarization and neutrophil activation were measured by flow cytometry. RNA sequencing (RNA-seq) was applied to screen for differentially expressed genes (DEGs) in lung tissues. RT-qPCR and Western blot assays were utilized to validate key DEGs and target proteins in lung tissues. 16S rRNA sequencing was employed to characterize the gut microbiota of mice. Metabolites in the gut were analyzed using untargeted metabolomics. RESULTS: FZQS treatment significantly ameliorated lung histopathological damage, decreased pro-inflammatory cytokine levels, and increased anti-inflammatory cytokine levels. M1 macrophage levels in the peripheral blood decreased, M2 macrophage levels increased, and activated neutrophils were inhibited in mice with LPS-induced lung injury. Importantly, transcriptomic analysis showed that FZQS downregulated macrophage and neutrophil activation and migration and adhesion pathways by reversing 51 DEGs, which was further confirmed by RT-qPCR and Western blot analysis. In addition, FZQS modulated the dysbiosis of the gut microbiota by reversing the abundance of Corynebacterium, Facklamia, Staphylococcus, Paenalcaligenes, Lachnoclostridium, norank_f_Muribaculaceae, and unclassified_f_Lachnospiraceae. Meanwhile, metabolomics analysis revealed that FZQS significantly regulated tryptophan metabolism by reducing the levels of 3-Indoleacetonitrile and 5-Hydroxykynurenine. CONCLUSION: FZQS effectively ameliorated LPS-induced lung injury by inhibiting the activation, migration, and adhesion of macrophages and neutrophils and modulating gut microbiota and its metabolites.


Sujet(s)
Médicaments issus de plantes chinoises , Lipopolysaccharides , Lésion pulmonaire , Métabolomique , Animaux , Médicaments issus de plantes chinoises/pharmacologie , Souris , Lésion pulmonaire/traitement médicamenteux , Lésion pulmonaire/métabolisme , Lésion pulmonaire/induit chimiquement , Mâle , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Transcriptome/effets des médicaments et des substances chimiques , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Souris de lignée C57BL , Cytokines/métabolisme , Modèles animaux de maladie humaine , Lésion pulmonaire aigüe/traitement médicamenteux , Lésion pulmonaire aigüe/induit chimiquement , Lésion pulmonaire aigüe/métabolisme
19.
J Investig Med High Impact Case Rep ; 12: 23247096241266089, 2024.
Article de Anglais | MEDLINE | ID: mdl-39051455

RÉSUMÉ

Pulmonary contusion (PC), defined as damage to the lung parenchyma with edema and hemorrhage, has classically been associated with acceleration-deceleration injuries. It is a frequent pathology in clinical practice. However, its clinical presentation and imaging findings are nonspecific. Patients with this entity can present with findings that can range from mild dyspnea to life-threatening respiratory failure and hemodynamic instability. We present the case of a 61-year-old man, a former smoker, who presented to the emergency department after suffering blunt chest trauma. On admission, he complained of only mild shortness of breath, and his vital signs were typical. Initial imaging identified asymmetric pulmonary infiltrates and mediastinal lymphadenopathy; this was suspicious for additional pathology in addition to PC. After an exhaustive evaluation, a neoplastic or infectious disease process was ruled out. Even though the patient presented with a clinical deterioration of respiratory function compatible with secondary acute respiratory distress syndrome, there was a complete recovery after supportive measures and supplemental oxygen. In conclusion, the nonspecific clinical and imaging findings in patients with pulmonary contusion warrant a complete evaluation of these cases. An early diagnosis is essential to establish adequate support and monitoring to prevent possible complications that could worsen the patient's prognosis.


Sujet(s)
Contusions , Lésion pulmonaire , Plaies non pénétrantes , Humains , Mâle , Adulte d'âge moyen , Contusions/imagerie diagnostique , Lésion pulmonaire/imagerie diagnostique , Lésion pulmonaire/étiologie , Plaies non pénétrantes/complications , Plaies non pénétrantes/imagerie diagnostique , /étiologie , /imagerie diagnostique , Tomodensitométrie , Poumon/imagerie diagnostique , Dyspnée/étiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE