RÉSUMÉ
Brain death (BD) provides most of the donor organs destined for lung transplantation (LTx). However, the organs may be affected by inflammatory and oxidative processes. Based on this, we hypothesize that the angiotensin-converting enzyme 2 (ACE2) activation can reduce the lung injury associated with LTx. 3 h after BD induction, rats were injected with saline (BD group) or an ACE2 activator (ACE2a group; 15 mg/kg-1) and kept on mechanical ventilation for additional 3 h. A third group included a control ventilation (Control group) prior to transplant. After BD protocol, left LTx were performed, followed by 2 h-reperfusion. ACE2 activation was associated with better oxygenation after BD management (p = 0.01), attenuating edema (p = 0.05) followed by the reduction in tissue resistance (p = 0.01) and increase of respiratory compliance (p = 0.02). Nrf2 expression was also upregulated in the ACE2a group (p = 0.03). After transplantation, ACE2a group showed lower levels of TNF-α (p = 0.02), IL-6 (p = 0.001), IL-1ß (p = 0.01), ROS (p = 0.004) and MDA (p = 0.002), in addition to higher CAT activity (p = 0.04). In conclusion, our study suggests that ACE2 activation improves anti-inflammatory and antioxidant activity in a model of LTx.
Sujet(s)
Angiotensin-converting enzyme 2 , Mort cérébrale , Inflammation , Transplantation pulmonaire , Stress oxydatif , Animaux , Angiotensin-converting enzyme 2/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Transplantation pulmonaire/effets indésirables , Rats , Inflammation/métabolisme , Mâle , Peptidyl-Dipeptidase A/métabolisme , Donneurs de tissus , Poumon/métabolisme , Poumon/anatomopathologieRÉSUMÉ
The developmental origins of healthy and disease (DOHaD) concept has demonstrated a higher rate of chronic diseases in the adult population of individuals whose mothers experienced severe maternal protein restriction (MPR). Using proteomic and in silico analyses, we investigated the lung proteomic profile of young and aged rats exposed to MPR during pregnancy and lactation. Our results demonstrated that MPR lead to structural and immune system pathways changes, and this outcome is coupled with a rise in the PI3k-AKT-mTOR signaling pathway, with increased MMP-2 activity, and CD8 expression in the early life, with long-term effects with aging. This led to the identification of commonly or inversely differentially expressed targets in early life and aging, revealing dysregulated pathways related to the immune system, stress, muscle contraction, tight junctions, and hemostasis. We identified three miRNAs (miR-378a-3p, miR-378a-5p, let-7a-5p) that regulate four proteins (ACTN4, PPIA, HSPA5, CALM1) as probable epigenetic lung marks generated by MPR. In conclusion, MPR impacts the lungs early in life, increasing the possibility of long-lasting negative outcomes for respiratory disorders in the offspring.
Sujet(s)
Poumon , microARN , Protéomique , Animaux , Femelle , Poumon/métabolisme , Mâle , Protéomique/méthodes , Grossesse , microARN/génétique , microARN/métabolisme , Rats , Effets différés de l'exposition prénatale à des facteurs de risque/métabolisme , Effets différés de l'exposition prénatale à des facteurs de risque/génétique , Régime pauvre en protéines , Transduction du signal , Sérine-thréonine kinases TOR/métabolisme , Sérine-thréonine kinases TOR/génétique , Longévité/génétique , Rat Wistar , Protéines proto-oncogènes c-akt/métabolisme , Protéome/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/génétique , Vieillissement/métabolisme , Vieillissement/génétique , Matrix metalloproteinase 2/métabolisme , Matrix metalloproteinase 2/génétiqueRÉSUMÉ
Several lung diseases can cause structural damage, making lung transplantation the only therapeutic option for advanced disease stages. However, the transplantation success rate remains limited. Lung bioengineering using the natural extracellular matrix (ECM) of decellularized lungs is a potential alternative. The use of undifferentiated cells to seed the ECM is practical; however, sterilizing the organ for recellularization is challenging. Photobiomodulation therapy (PBMT) may offer a solution, in which the wavelength is crucial for tissue penetration. This study aimed to explore the potential of optimizing lung recellularization with mesenchymal stem cells using PBMT (660 nm) after sterilization with PBMT (880 nm). The lungs from C57BL/6 mice were decellularized using 1% SDS and sterilized using PBMT (880 nm, 100 mW, 30 s). Recellularization was performed in two groups: (1) recellularized lung and (2) recellularized lung + 660 nm PBMT (660 nm, 100 mW, 30 s). Both were seeded with mesenchymal stem cells from human tooth pulp (DPSc) and incubated for 24 h at 37 °C and 5% CO2 in bioreactor-like conditions with continuous positive airway pressure (CPAP) at 20 cmH2O and 90% O2. The culture medium was analyzed after 24 h. H&E, immunostaining, SEM, and ELISA assays were performed. Viable biological scaffolds were produced, which were free of cell DNA and preserved the glycosaminoglycans; collagens I, III, and IV; fibronectin; laminin; elastin; and the lung structure (SEM). The IL-6 and IL-8 levels were stable during the 24 h culture, but the IFN-γ levels showed significant differences in the recellularized lung and recellularized lung + 660 nm PBMT groups. Greater immunological modulation was observed in the recellularized groups regarding pro-inflammatory cytokines (IL-6, IFN-γ, and IL-8). These findings suggest that PBMT plays a role in cytokine regulation and antimicrobial activity, thus offering promise for enhanced therapeutic strategies in lung bioengineering.
Sujet(s)
Cytokines , Photothérapie de faible intensité , Poumon , Cellules souches mésenchymateuses , Souris de lignée C57BL , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/métabolisme , Animaux , Souris , Poumon/métabolisme , Photothérapie de faible intensité/méthodes , Humains , Cytokines/métabolisme , Transplantation de cellules souches mésenchymateuses/méthodes , Stérilisation/méthodes , Matrice extracellulaire/métabolisme , Ingénierie tissulaire/méthodesRÉSUMÉ
Pseudomonas aeruginosa is a frequent cause of antimicrobial-resistant hospital-acquired pneumonia, especially in critically ill patients. Inflammation triggered by P. aeruginosa infection is necessary for bacterial clearance but must be spatially and temporally regulated to prevent further tissue damage and bacterial dissemination. Emerging data have shed light on the pro-resolving actions of angiotensin-(1-7) [Ang-(1-7)] signaling through the G protein-coupled receptor Mas (MasR) during infections. Herein, we investigated the role of the Ang-(1-7)/Mas axis in pneumonia caused by P. aeruginosa by using genetic and pharmacological approach and found that Mas receptor-deficient animals developed a more severe form of pneumonia showing higher neutrophilic infiltration into the airways, bacterial load, cytokines, and chemokines production and more severe pulmonary damage. Conversely, treatment of pseudomonas-infected mice with Ang-(1-7) was able to decrease neutrophilic infiltration in airways and lungs, local and systemic levels of pro-inflammatory cytokines and chemokines, and increase the efferocytosis rates, mitigating lung damage/dysfunction caused by infection. Notably, the therapeutic association of Ang-(1-7) with antibiotics improved the survival rates of mice subjected to lethal inoculum of P. aeruginosa, extending the therapeutic window for imipenem. Mechanistically, Ang-(1-7) increased phagocytosis of bacteria by neutrophils and macrophages to accelerate pathogen clearance. Altogether, harnessing the Ang-(1-7) pathway during infection is a potential strategy for the development of host-directed therapies to promote mechanisms of resistance and resilience to pneumonia.
Sujet(s)
Angiotensine-I , Antibactériens , Souris de lignée C57BL , Fragments peptidiques , Proto-oncogène Mas , Infections à Pseudomonas , Pseudomonas aeruginosa , Récepteurs couplés aux protéines G , Animaux , Angiotensine-I/métabolisme , Pseudomonas aeruginosa/effets des médicaments et des substances chimiques , Souris , Infections à Pseudomonas/traitement médicamenteux , Infections à Pseudomonas/métabolisme , Infections à Pseudomonas/microbiologie , Fragments peptidiques/métabolisme , Fragments peptidiques/pharmacologie , Récepteurs couplés aux protéines G/métabolisme , Antibactériens/pharmacologie , Antibactériens/usage thérapeutique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Pneumopathie bactérienne/traitement médicamenteux , Pneumopathie bactérienne/microbiologie , Pneumopathie bactérienne/anatomopathologie , Pneumopathie bactérienne/métabolisme , Cytokines/métabolisme , Souris knockout , Pneumopathie infectieuse/traitement médicamenteux , Pneumopathie infectieuse/métabolisme , Pneumopathie infectieuse/microbiologie , Mâle , Poumon/microbiologie , Poumon/métabolisme , Poumon/anatomopathologie , Transduction du signal/effets des médicaments et des substances chimiques , Infiltration par les neutrophiles/effets des médicaments et des substances chimiquesRÉSUMÉ
Omega-3 (ω3) fatty acids are widely investigated for their anti-inflammatory potential, however, there is little evidence regarding their action in the lung parenchyma in the context of obesity. The objective is to investigate the effects of flaxseed oil (FS), rich in α-linolenic (C18:3 - ω3), on the lungs of obese mice. Mice were fed a high-fat diet (HF) for 8 weeks to induce obesity. Subsequently, a part of these animals received HF containing FS oil for another 8 weeks. The HF consumption induced weight gain and hyperglycemia. The lung parenchyma shows a complete fatty acids profile, compared to the control group (CT). In the lung parenchyma, FS increases the ω3 content and, notwithstanding a reduction in the interleukins (IL) IL1ß and IL18 contents compared to HF. However, FS promoted increased alveolar spaces, followed by MCP1 (Monocytes Chemoattractant Protein-1) positive cell infiltration and a dramatic reduction in the anti-inflammatory cytokine, IL10. Despite reducing the pulmonary inflammatory response, the consumption of a food source of ω3 was associated with alterations in the lipid profile and histoarchitecture of the lung parenchyma, which can lead to the development of pulmonary complications. This study brings an alert against the indiscriminate use of ω3 supplements, warranting caution.
Sujet(s)
Alimentation riche en graisse , Huile de lin , Poumon , Obésité , Animaux , Huile de lin/pharmacologie , Huile de lin/administration et posologie , Alimentation riche en graisse/effets indésirables , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Obésité/diétothérapie , Obésité/métabolisme , Mâle , Souris de lignée C57BL , Souris , Souris obèse , Interleukine-1 bêta/métabolisme , Interleukine-18/métabolisme , Chimiokine CCL2/métabolisme , Interleukine-10/métabolisme , Acides gras omega-3/pharmacologie , Acides gras omega-3/administration et posologie , Prise de poids/effets des médicaments et des substances chimiquesRÉSUMÉ
Ash emission from volcanic eruptions affects the environment, society, and human health. This study shows the total concentration and lung bioaccessible fraction of eight potential toxic metal(loid)s in five Popocatépetl ashfall samples. Mineralogical phases and particle size distribution of the ashfall were analyzed by X-ray diffraction (XRD) and Scanning Electron Microscope (SEM) techniques, respectively. The bioaccessibility test of Gamble solution (GS) and Artificial Lysosomal Fluid (ALF) were conducted to simulate extracellular (pH 7) and intracellular (pH 4.5) conditions, respectively. The studied metal(loid)s showed the following total concentration (mg kg-1): 1.98 (As), 0.17 (Cd), 134.09 (Cr), 8.66 (Cu), 697.33 (Mn), 55.35 (Ni), 8.77 (Pb), and 104.10 (Zn). Geochemical indices suggested that some metal(loid)s are slightly enriched compared to the local soil background concentrations. Several mineralogical phases were identified in the collected ashfall deposits, such as plagioclase, pyroxene, and Fe-Ti oxide, among others. According to the risk assessment results, the non-carcinogenic risk related to ashfall exposure returns an HQ > 1 for children. In contrast, the estimation of carcinogenic risk was found to be within the tolerable limit. Metal(loid)s showed low bioaccessibility (< 30%) in GS and ALF, with the highest values found in ALF solution for As (12.18%) and Cu (7.57%). Despite their metal-bioaccessibility, our findings also showed that dominant ash particle size ranged between fine (< 2.5 µm) and extremely fine (< 1 µm), considered highly inhalable fractions. The results obtained in this work indicate that volcanic ashes are bioinsoluble and biodurable, and exhibit low bioaccessibility when in contact with lung human fluids.
Sujet(s)
Éruptions volcaniques , Appréciation des risques , Humains , Mexique , Biodisponibilité , Taille de particule , Poumon/métabolisme , Poumon/composition chimique , Surveillance de l'environnement/méthodes , Microscopie électronique à balayage , Métaux/analyse , Diffraction des rayons X , Exposition environnementale , Polluants atmosphériques/analyseRÉSUMÉ
Asthma is a chronic immunological disease related to oxidative stress and chronic inflammation; both processes promote airway remodeling with collagen deposition and matrix thickening, causing pulmonary damage and lost function. This study investigates the immunomodulation of C-phycocyanin (CPC), a natural blue pigment purified from cyanobacteria, as a potential alternative treatment to prevent the remodeling process against asthma. We conducted experiments using ovalbumin (OVA) to induce asthma in Sprague Dawley rats. Animals were divided into five groups: (1) sham + vehicle, (2) sham + CPC, (3) asthma + vehicle, (4) asthma + CPC, and (5) asthma + methylprednisolone (MP). Our findings reveal that asthma promotes hypoxemia, leukocytosis, and pulmonary myeloperoxidase (MPO) activity by increasing lipid peroxidation, reactive oxygen and nitrogen species, inflammation associated with Th2 response, and airway remodeling in the lungs. CPC and MP treatment partially prevented these physiological processes with similar action on the biomarkers evaluated. In conclusion, CPC treatment enhanced the antioxidant defense system, thereby preventing oxidative stress and reducing airway inflammation by regulating pro-inflammatory and anti-inflammatory cytokines, consequently avoiding asthma-induced airway remodeling.
Sujet(s)
Remodelage des voies aériennes , Asthme , Modèles animaux de maladie humaine , Ovalbumine , Stress oxydatif , Phycocyanine , Rat Sprague-Dawley , Animaux , Phycocyanine/pharmacologie , Phycocyanine/usage thérapeutique , Asthme/traitement médicamenteux , Asthme/métabolisme , Asthme/induit chimiquement , Stress oxydatif/effets des médicaments et des substances chimiques , Ovalbumine/effets indésirables , Rats , Remodelage des voies aériennes/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Mâle , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Cytokines/métabolismeRÉSUMÉ
The combination of a polyphenol, quercetin, with dasatinib initiated clinical trials to evaluate the safety and efficacy of senolytics in idiopathic pulmonary fibrosis, a lung disease associated with the presence of senescent cells. Another approach to senotherapeutics consists of controlling inflammation related to cellular senescence or "inflammaging", which participates, among other processes, in establishing pulmonary fibrosis. We evaluate whether polyphenols such as caffeic acid, chlorogenic acid, epicatechin, gallic acid, quercetin, or resveratrol combined with different senotherapeutics such as metformin or rapamycin, and antifibrotic drugs such as nintedanib or pirfenidone, could present beneficial actions in an in vitro model of senescent MRC-5 lung fibroblasts. A senescent-associated secretory phenotype (SASP) was evaluated by the measurement of interleukin (IL)-6, IL-8, and IL-1ß. The senescent-associated ß-galactosidase (SA-ß-gal) activity and cellular proliferation were assessed. Fibrosis was evaluated using a Picrosirius red assay and the gene expression of fibrosis-related genes. Epithelial-mesenchymal transition (EMT) was assayed in the A549 cell line exposed to Transforming Growth Factor (TGF)-ß in vitro. The combination that demonstrated the best results was metformin and caffeic acid, by inhibiting IL-6 and IL-8 in senescent MRC-5 cells. Metformin and caffeic acid also restore cellular proliferation and reduce SA-ß-gal activity during senescence induction. The collagen production by senescent MRC-5 cells was inhibited by epicatechin alone or combined with drugs. Epicatechin and nintedanib were able to control EMT in A549 cells. In conclusion, caffeic acid and epicatechin can potentially increase the effectiveness of senotherapeutic drugs in controlling lung diseases whose pathophysiological component is the presence of senescent cells and fibrosis.
Sujet(s)
Vieillissement de la cellule , Fibroblastes , Poumon , Polyphénols , Humains , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Polyphénols/pharmacologie , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Cellules A549 , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Metformine/pharmacologie , Acides caféiques/pharmacologie , Indoles/pharmacologie , Sénothérapie/pharmacologie , Lignée cellulaire , Phénotype sécrétoire associé à la sénescence/effets des médicaments et des substances chimiques , Sirolimus/pharmacologie , Interleukine-8/métabolisme , Interleukine-8/génétique , Facteur de croissance transformant bêta/métabolisme , PyridonesRÉSUMÉ
Currently, the global lifespan has increased, resulting in a higher proportion of the population over 65 years. Changes that occur in the lung during aging increase the risk of developing acute and chronic lung diseases, such as acute respiratory distress syndrome, chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, and lung cancer. During normal tissue homeostasis, cell proliferation and apoptosis create a dynamic balance that constitutes the physiological cell turnover. In basal conditions, the lungs have a low rate of cell turnover compared to other organs. During aging, changes in the rate of cell turnover in the lung are observed. In this work, we review the literature that evaluates the role of molecules involved in cell proliferation and apoptosis in lung aging and in the development of age-related lung diseases. The list of molecules that regulate cell proliferation, apoptosis, or both processes in lung aging includes TNC, FOXM1, DNA-PKcs, MicroRNAs, BCL-W, BCL-XL, TCF21, p16, NOX4, NRF2, MDM4, RPIA, DHEA, and MMP28. However, despite the studies carried out to date, the complete signaling pathways that regulate cell turnover in lung aging are still unknown. More research is needed to understand the changes that lead to the development of age-related lung diseases.
Sujet(s)
Vieillissement , Apoptose , Prolifération cellulaire , Poumon , Humains , Vieillissement/physiologie , Poumon/métabolisme , Poumon/anatomopathologie , Animaux , Transduction du signal , Maladies pulmonaires/anatomopathologie , Maladies pulmonaires/métabolismeRÉSUMÉ
Neurotoxicity is a major obstacle in the effectiveness of Cisplatin in cancer chemotherapy. In this process, oxidative stress and inflammation are considered to be the main mechanisms involved in brain and lung toxicity. The aim of the present work was to study the influence of the amount of protein on some oxidative parameters in the brain and lungs of rats treated with Cisplatin (CP) and N-Acetylcysteine (NAC) as neuroprotectors. Four groups of Wistar rats, each containing six animals, were fed with a protein diet at 7% for 15 days. Thereafter, the groups were given either a unique dose of CP® 5 mg/kg or NAC® 5 mg/kg as follows: group 1 (control), NaCl 0.9% vehicle; group 2, CP; group 3, NAC; and group 4, NAC + CP. The animals were sacrificed immediately after the treatments. Blood samples were collected upon sacrifice and used to measure blood triglycerides and glucose. The brain and lungs of each animal were obtained and used to assay lipid peroxidation (TBARS), glutathione (GSH), serotonin metabolite (5-HIAA), catalase, and the activity of Ca+2, and Mg+2 ATPase using validated methods. TBARS, H2O2, and GSH were found to be significantly decreased in the cortex and cerebellum/medulla oblongata of the groups treated with CP and NAC. The total ATPase showed a significant increase in the lung and cerebellum/medulla oblongata, while 5-HIAA showed the same tendency in the cortex of the same group of animals. The increase in 5-HIAA and ATPase during NAC and CP administration resulted in brain protection. This effect could be even more powerful when membrane fluidity is increased, thus proving the efficacy of combined NAC and CP drug therapy, which appears to be a promising strategy for future chemotherapy in malnourished patients.
Sujet(s)
Acétylcystéine , Cisplatine , Poumon , Rat Wistar , Animaux , Cisplatine/effets indésirables , Cisplatine/toxicité , Acétylcystéine/pharmacologie , Rats , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Poumon/anatomopathologie , Peroxydation lipidique/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Mâle , Cerveau/effets des médicaments et des substances chimiques , Cerveau/métabolisme , Glutathion/métabolisme , Neuroprotecteurs/pharmacologie , Antinéoplasiques/effets indésirablesRÉSUMÉ
This study introduces the nanobromhexine lipid particle (NBL) platform designed for effective pulmonary drug delivery. Inspired by respiratory virus transport mechanisms, NBL address challenges associated with mucus permeation and inflammation in pulmonary diseases. Composed of low molecular weight polyethylene glycol-coated lipid nanoparticles with bromhexine hydrochloride, NBL exhibit a size of 118 ± 24 nm, a neutral zeta potential, osmolarity of 358 ± 28 mOsmol/kg, and a pH of 6.5. Nebulizing without leakage and showing no toxicity to epithelial cells, NBL display mucoadhesive properties with a 60% mucin-binding efficiency. They effectively traverse the dense mucus layer of Calu-3 cultures in an air-liquid interface, as supported by a 55% decrease in MUC5AC density and a 29% increase in nanoparticles internalization compared to non-exposed cells. In assessing immunomodulatory effects, NBL treatment in SARS-CoV-2-infected lung cells leads to a 40-fold increase in anti-inflammatory MUC1 gene expression, a proportional reduction in pro-inflammatory IL-6 expression, and elevated anti-inflammatory IL-10 expression. These findings suggest a potential mechanism to regulate the excessive IL-6 expression triggered by virus infection. Therefore, the NBL platform demonstrates promising potential for efficient pulmonary drug delivery and immunomodulation, offering a novel approach to addressing mucus permeation and inflammation in pulmonary diseases.
Sujet(s)
Poumon , Nanoparticules , SARS-CoV-2 , Nanoparticules/administration et posologie , Humains , Poumon/métabolisme , SARS-CoV-2/effets des médicaments et des substances chimiques , Systèmes de délivrance de médicaments , Immunomodulation , Lignée cellulaire , Mucine-1/métabolisme , COVID-19 , Lipides/composition chimique , Lipides/administration et posologie , Mucus/métabolisme , Polyéthylène glycols/composition chimique , Cellules épithéliales/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Traitements médicamenteux de la COVID-19 , Mucine-5AC/métabolisme , LiposomesRÉSUMÉ
Endothelial glycocalyx (eGC) covers the inner surface of the vessels and plays a role in vascular homeostasis. Syndecan is considered the "backbone" of this structure. Several studies have shown eGC shedding in sepsis and its involvement in organ dysfunction. Matrix metalloproteinases (MMP) contribute to eGC shedding through their ability for syndecan-1 cleavage. This study aimed to investigate if doxycycline, a potent MMP inhibitor, could protect against eGC shedding in lipopolysaccharide (LPS)-induced sepsis and if it could interrupt the vascular hyperpermeability, neutrophil transmigration, and microvascular impairment. Rats that received pretreatment with doxycycline before LPS displayed ultrastructural preservation of the eGC observed using transmission electronic microscopy of the lung and heart. In addition, these animals exhibited lower serum syndecan-1 levels, a biomarker of eGC injury, and lower perfused boundary region (PBR) in the mesenteric video capillaroscopy, which is inversely related to the eGC thickness compared with rats that only received LPS. Furthermore, this study revealed that doxycycline decreased sepsis-related vascular hyperpermeability in the lung and heart, reduced neutrophil transmigration in the peritoneal lavage and inside the lungs, and improved some microvascular parameters. These findings suggest that doxycycline protects against LPS-induced eGC shedding, and it could reduce vascular hyperpermeability, neutrophils transmigration, and microvascular impairment.
Sujet(s)
Doxycycline , Glycocalyx , Lipopolysaccharides , Sepsie , Glycocalyx/métabolisme , Glycocalyx/effets des médicaments et des substances chimiques , Animaux , Sepsie/traitement médicamenteux , Sepsie/métabolisme , Doxycycline/pharmacologie , Rats , Mâle , Perméabilité capillaire/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Poumon/effets des médicaments et des substances chimiques , Syndécane-1/métabolisme , Rat Wistar , Endothélium vasculaire/effets des médicaments et des substances chimiques , Endothélium vasculaire/métabolisme , Endothélium vasculaire/anatomopathologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Inhibiteurs de métalloprotéinases matricielles/pharmacologieRÉSUMÉ
Introduction: TAM receptor-mediated efferocytosis plays an important function in immune regulation and may contribute to antigen tolerance in the lungs, a site with continuous cellular turnover and generation of apoptotic cells. Some studies have identified failures in efferocytosis as a common driver of inflammation and tissue destruction in lung diseases. Our study is the first to characterize the in vivo function of the TAM receptors, Axl and MerTk, in the innate immune cell compartment, cytokine and chemokine production, as well as the alveolar macrophage (AM) phenotype in different settings in the airways and lung parenchyma. Methods: We employed MerTk and Axl defective mice to induce acute silicosis by a single exposure to crystalline silica particles (20 mg/50 µL). Although both mRNA levels of Axl and MerTk receptors were constitutively expressed by lung cells and isolated AMs, we found that MerTk was critical for maintaining lung homeostasis, whereas Axl played a role in the regulation of silica-induced inflammation. Our findings imply that MerTk and Axl differently modulated inflammatory tone via AM and neutrophil recruitment, phenotype and function by flow cytometry, and TGF-ß and CXCL1 protein levels, respectively. Finally, Axl expression was upregulated in both MerTk-/- and WT AMs, confirming its importance during inflammation. Conclusion: This study provides strong evidence that MerTk and Axl are specialized to orchestrate apoptotic cell clearance across different circumstances and may have important implications for the understanding of pulmonary inflammatory disorders as well as for the development of new approaches to therapy.
Sujet(s)
Axl Receptor Tyrosine Kinase , Homéostasie , Poumon , Macrophages alvéolaires , Souris knockout , Protéines proto-oncogènes , Récepteurs à activité tyrosine kinase , Silicose , c-Mer Tyrosine kinase , Animaux , Souris , c-Mer Tyrosine kinase/métabolisme , c-Mer Tyrosine kinase/génétique , Cytokines/métabolisme , Modèles animaux de maladie humaine , Poumon/immunologie , Poumon/métabolisme , Poumon/anatomopathologie , Macrophages alvéolaires/immunologie , Macrophages alvéolaires/métabolisme , Souris de lignée C57BL , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Récepteurs à activité tyrosine kinase/métabolisme , Récepteurs à activité tyrosine kinase/génétique , Silicose/métabolisme , Silicose/immunologie , Silicose/anatomopathologie , MâleRÉSUMÉ
BACKGROUND: Cytokine storm and oxidative stress are present in chronic obstructive pulmonary disease (COPD). Individuals with COPD present high levels of NF-κB-associated cytokines and pro-oxidant agents as well as low levels of Nrf2-associated antioxidants. This condition creates a steroid-resistant inflammatory microenvironment. Lacticaseibacillus rhamnosus (Lr) is a known anti-cytokine in lung diseases; however, the effect of Lr on lung inflammation and oxidative stress in steroid-resistant COPD mice remains unknown. OBJECTIVE: Thus, we investigated the Lr effect on lung inflammation and oxidative stress in mice and macrophages exposed to cigarette smoke extract (CSE) and unresponsive to steroids. METHODS: Mice and macrophages received dexamethasone or GLPG-094 (a GPR43 inhibitor), and only the macrophages received butyrate (but), all treatments being given before CSE. Lung inflammation was evaluated from the leukocyte population, airway remodeling, cytokines, and NF-κB. Oxidative stress disturbance was measured from ROS, 8-isoprostane, NADPH oxidase, TBARS, SOD, catalase, HO-1, and Nrf2. RESULTS: Lr attenuated cellularity, mucus, collagen, cytokines, ROS, 8-isoprostane, NADPH oxidase, and TBARS. Otherwise, SOD, catalase, HO-1, and Nrf2 were upregulated in Lr-treated COPD mice. Anti-cytokine and antioxidant effects of butyrate also occurred in CSE-exposed macrophages. GLPG-094 rendered Lr and butyrate less effective. CONCLUSIONS: Lr attenuates lung inflammation and oxidative stress in COPD mice, suggesting the presence of a GPR43 receptor-dependent mechanism also found in macrophages.
Sujet(s)
Lacticaseibacillus rhamnosus , Macrophages , Stress oxydatif , Broncho-pneumopathie chronique obstructive , Récepteurs couplés aux protéines G , Animaux , Broncho-pneumopathie chronique obstructive/traitement médicamenteux , Broncho-pneumopathie chronique obstructive/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Récepteurs couplés aux protéines G/métabolisme , Souris , Humains , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Mâle , Cytokines/métabolisme , Médiateurs de l'inflammation/métabolisme , Souris de lignée C57BL , Modèles animaux de maladie humaine , Fumée/effets indésirables , Dexaméthasone/pharmacologie , Butyrates/pharmacologie , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolismeRÉSUMÉ
Introduction: Brain death (BD) is known to compromise graft quality by causing hemodynamic, metabolic, and hormonal changes. The abrupt reduction of female sex hormones after BD was associated with increased lung inflammation. The use of both corticoids and estradiol independently has presented positive results in modulating BD-induced inflammatory response. However, studies have shown that for females the presence of both estrogen and corticoids is necessary to ensure adequate immune response. In that sense, this study aims to investigate how the association of methylprednisolone (MP) and estradiol (E2) could modulate the lung inflammation triggered by BD in female rats. Methods: Female Wistar rats (8 weeks) were divided into four groups: sham (animals submitted to the surgical process, without induction of BD), BD (animals submitted to BD), MP/E2 (animals submitted to BD that received MP and E2 treatment 3h after BD induction) and MP (animals submitted to BD that received MP treatment 3h after BD induction). Results: Hemodynamics, systemic and local quantification of IL-6, IL-1ß, VEGF, and TNF-α, leukocyte infiltration to the lung parenchyma and airways, and adhesion molecule expression were analyzed. After treatment, MP/E2 association was able to reinstate mean arterial pressure to levels close to Sham animals (p<0.05). BD increased leukocyte infiltration to the airways and MP/E2 was able to reduce the number of cells (p=0.0139). Also, the associated treatment modulated the vasculature by reducing the expression of VEGF (p=0.0616) and maintaining eNOS levels (p=0.004) in lung tissue. Discussion: Data presented in this study show that the association between corticoids and estradiol could represent a better treatment strategy for lung inflammation in the female BD donor by presenting a positive effect in the hemodynamic management of the donor, as well as by reducing infiltrated leukocyte to the airways and release of inflammatory markers in the short and long term.
Sujet(s)
Mort cérébrale , Oestradiol , Méthylprednisolone , Pneumopathie infectieuse , Rat Wistar , Animaux , Femelle , Oestradiol/pharmacologie , Méthylprednisolone/pharmacologie , Rats , Pneumopathie infectieuse/traitement médicamenteux , Pneumopathie infectieuse/métabolisme , Cytokines/métabolisme , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Poumon/immunologie , Modèles animaux de maladie humaine , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutiqueRÉSUMÉ
ABSTRACT: Myocardial infarction (MI) and pulmonary arterial hypertension (PAH) are 2 prevalent cardiovascular diseases. In both conditions, oxidative stress is associated with a worse prognosis. Pterostilbene (PTE), an antioxidant compound, has been studied as a possible therapy for cardiovascular diseases. This study aims to evaluate the effect of PTE on oxidative stress in the hearts of animals with MI and in the lungs of animals with PAH. Male Wistar rats were used in both models. In the MI model, the experimental groups were sham, MI, and MI + PTE. In the PAH model, the experimental groups were control, PAH, and PAH + PTE. Animals were exposed to MI through surgical ligation of the left coronary artery, or to PAH, by the administration of monocrotaline (60 mg/kg). Seven days after undergoing cardiac injury, the MI + PTE animals were treated with PTE (100 mg/kg day) for 8 days. After this, the heart was collected for molecular analysis. The PAH + PTE animals were treated with PTE (100 mg/kg day) for 14 days, beginning 7 days after PAH induction. After this, the lungs were collected for biochemical evaluation. We found that PTE administration attenuated the decrease in ejection fraction and improved left ventricle end-systolic volume in infarcted animals. In the PAH model, PTE improved pulmonary artery flow and decreased reactive oxygen species levels in the lung. PTE administration promoted protective effects in terms of oxidative stress in 2 experimental models of cardiac diseases: MI and PAH. PTE also improved cardiac function in infarcted rats and pulmonary artery flow in animals with PAH.
Sujet(s)
Antioxydants , Modèles animaux de maladie humaine , Poumon , Infarctus du myocarde , Myocarde , Stress oxydatif , Hypertension artérielle pulmonaire , Rat Wistar , Stilbènes , Animaux , Stress oxydatif/effets des médicaments et des substances chimiques , Mâle , Infarctus du myocarde/physiopathologie , Infarctus du myocarde/métabolisme , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Poumon/physiopathologie , Stilbènes/pharmacologie , Hypertension artérielle pulmonaire/traitement médicamenteux , Hypertension artérielle pulmonaire/physiopathologie , Hypertension artérielle pulmonaire/métabolisme , Antioxydants/pharmacologie , Myocarde/métabolisme , Myocarde/anatomopathologie , Artère pulmonaire/effets des médicaments et des substances chimiques , Artère pulmonaire/physiopathologie , Artère pulmonaire/métabolisme , Fonction ventriculaire gauche/effets des médicaments et des substances chimiques , Rats , Espèces réactives de l'oxygène/métabolisme , Pression artérielle/effets des médicaments et des substances chimiques , MonocrotalineRÉSUMÉ
Prolonged exposure to iron powder and other mineral dusts can threaten the health of individuals, especially those with COPD. The goal of this study was to determine how environmental exposure to metal dust from two different mining centers in Brazil affects lung mechanics, inflammation, remodeling and oxidative stress responses in healthy and elastase-exposed mice. This study divided 72 male C57Bl/6 mice into two groups, the summer group and the winter group. These groups were further divided into six groups: control, nonexposed (SAL); nonexposed, given elastase (ELA); exposed to metal powder at a mining company (SAL-L1 and ELA-L1); and exposed to a location three miles away from the mining company (SAL-L2 and ELA-L2) for four weeks. On the 29th day of the protocol, the researchers assessed lung mechanics, bronchoalveolar lavage fluid (BALF), inflammation, remodeling, oxidative stress, macrophage iron and alveolar wall alterations (mean linear intercept-Lm). The Lm was increased in the ELA, ELA-L1 and ELA-L2 groups compared to the SAL group (p < 0.05). There was an increase in the total number of cells and macrophages in the ELA-L1 and ELA-L2 groups compared to the other groups (p < 0.05). Compared to the ELA and SAL groups, the exposed groups (ELA-L1, ELA-L2, SAL-L1, and SAL-L2) exhibited increased expression of IL-1ß, IL-6, IL-10, IL-17, TNF-α, neutrophil elastase, TIMP-1, MMP-9, MMP-12, TGF-ß, collagen fibers, MUC5AC, iNOS, Gp91phox, NFkB and iron positive macrophages (p < 0.05). Although we did not find differences in lung mechanics across all groups, there were low to moderate correlations between inflammation remodeling, oxidative stress and NFkB with elastance, resistance of lung tissue and iron positive macrophages (p < 0.05). Environmental exposure to iron, confirmed by evaluation of iron in alveolar macrophages and in air, exacerbated inflammation, initiated remodeling, and induced oxidative stress responses in exposed mice with and without emphysema. Activation of the iNOS, Gp91phox and NFkB pathways play a role in these changes.
Sujet(s)
Exposition environnementale , Fer , Pancreatic elastase , Animaux , Mâle , Souris , Liquide de lavage bronchoalvéolaire/composition chimique , Exposition environnementale/effets indésirables , Inflammation/métabolisme , Inflammation/induit chimiquement , Fer/toxicité , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Poumon/anatomopathologie , Souris de lignée C57BL , Stress oxydatif/effets des médicaments et des substances chimiques , Pancreatic elastase/métabolisme , Pancreatic elastase/pharmacologie , Poudres/toxicitéRÉSUMÉ
Introduction: Pulmonary diseases represent a significant burden to patients and the healthcare system and are one of the leading causes of mortality worldwide. Particularly, the COVID-19 pandemic has had a profound global impact, affecting public health, economies, and daily life. While the peak of the crisis has subsided, the global number of reported COVID-19 cases remains significantly high, according to medical agencies around the world. Furthermore, despite the success of vaccines in reducing the number of deaths caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), there remains a gap in the treatment of the disease, especially in addressing uncontrolled inflammation. The massive recruitment of leukocytes to lung tissue and alveoli is a hallmark factor in COVID-19, being essential for effectively responding to the pulmonary insult but also linked to inflammation and lung damage. In this context, mice models are a crucial tool, offering valuable insights into both the pathogenesis of the disease and potential therapeutic approaches. Methods: Here, we investigated the anti-inflammatory effect of the glycosaminoglycan (GAG)-binding chemokine fragment CXCL9(74-103), a molecule that potentially decreases neutrophil transmigration by competing with chemokines for GAG-binding sites, in two models of pneumonia caused by coronavirus infection. Results: In a murine model of betacoronavirus MHV-3 infection, the treatment with CXCL9(74-103) decreased the accumulation of total leukocytes, mainly neutrophils, to the alveolar space and improved several parameters of lung dysfunction 3 days after infection. Additionally, this treatment also reduced the lung damage. In the SARS-CoV-2 model in K18-hACE2-mice, CXCL9(74-103) significantly improved the clinical manifestations of the disease, reducing pulmonary damage and decreasing viral titers in the lungs. Discussion: These findings indicate that CXCL9(74-103) resulted in highly favorable outcomes in controlling pneumonia caused by coronavirus, as it effectively diminishes the clinical consequences of the infections and reduces both local and systemic inflammation.
Sujet(s)
COVID-19 , Chimiokine CXCL9 , Modèles animaux de maladie humaine , Glycosaminoglycanes , Poumon , SARS-CoV-2 , Animaux , Souris , COVID-19/immunologie , SARS-CoV-2/immunologie , Glycosaminoglycanes/métabolisme , Chimiokine CXCL9/métabolisme , Poumon/anatomopathologie , Poumon/virologie , Poumon/immunologie , Poumon/métabolisme , Inflammation/immunologie , Humains , Traitements médicamenteux de la COVID-19 , Souris de lignée C57BL , FemelleRÉSUMÉ
Severe cases of COVID-19 are characterized by development of acute respiratory distress syndrome (ARDS). Water accumulation in the lungs is thought to occur as consequence of an exaggerated inflammatory response. A possible mechanism could involve decreased activity of the epithelial Na+ channel, ENaC, expressed in type II pneumocytes. Reduced transepithelial Na+ reabsorption could contribute to lung edema due to reduced alveolar fluid clearance. This hypothesis is based on the observation of the presence of a novel furin cleavage site in the S protein of SARS-CoV-2 that is identical to the furin cleavage site present in the alpha subunit of ENaC. Proteolytic processing of αENaC by furin-like proteases is essential for channel activity. Thus, competition between S protein and αENaC for furin-mediated cleavage in SARS-CoV-2-infected cells may negatively affect channel activity. Here we present experimental evidence showing that coexpression of the S protein with ENaC in a cellular model reduces channel activity. In addition, we show that bidirectional competition for cleavage by furin-like proteases occurs between ãENaC and S protein. In transgenic mice sensitive to lethal SARS-CoV-2, however, a significant decrease in gamma ENaC expression was not observed by immunostaining of lungs infected as shown by SARS-CoV2 nucleoprotein staining.
Sujet(s)
COVID-19 , Canaux sodium épithéliaux , Furine , Souris transgéniques , Protéolyse , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Canaux sodium épithéliaux/métabolisme , Animaux , Humains , Souris , Furine/métabolisme , SARS-CoV-2/métabolisme , SARS-CoV-2/pathogénicité , Glycoprotéine de spicule des coronavirus/métabolisme , COVID-19/métabolisme , COVID-19/virologie , Pneumocytes/métabolisme , Pneumocytes/virologie , Poumon/métabolisme , Poumon/virologie , Poumon/anatomopathologie , Cellules HEK293RÉSUMÉ
Lung transplantation stands as a vital treatment for severe lung diseases, primarily sourcing organs from donors with brain death (BD). This research delved into the potential anti-inflammatory effects of thalidomide in rats with BD-induced lung complications. In this study twenty-four Wistar rats were divided into three groups: the control (CTR), brain death (BD) and brain death + thalidomide (TLD) groups. Post specific procedures, a 360 min monitoring period ensued. Comprehensive analyses of blood and heart-lung samples were conducted. Elevated IL-6 levels characterized both BD and TLD groups relative to the CTR (p = 0.0067 and p = 0.0137). Furthermore, TNF-α levels were notably higher in the BD group than both CTR and TLD (p = 0.0152 and p = 0.0495). Additionally, IL-1ß concentrations were significantly pronounced in both BD and TLD compared to CTR, with the BD group surpassing TLD (p = 0.0256). Immunohistochemical assessments revealed augmented NF-ĸB expression in the BD group in comparison to both CTR and TLD (p = 0.0006 and p = 0.0005). With this study we can conclude that BD induced acute pulmonary inflammation, whereas thalidomide manifested a notable capability in diminishing key inflammatory markers, indicating its prospective therapeutic significance in lung transplantation scenarios.