Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.113
Filter
1.
Sci Adv ; 10(34): eado8549, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39167644

ABSTRACT

Reduced skeletal muscle mass and oxidative capacity coexist in patients with pulmonary emphysema and are independently associated with higher mortality. If reduced cellular respiration contributes to muscle atrophy in that setting remains unknown. Using a mouse with genetically induced pulmonary emphysema that recapitulates muscle dysfunction, we found that reduced activity of succinate dehydrogenase (SDH) is a hallmark of its myopathic changes. We generated an inducible, muscle-specific SDH knockout mouse that demonstrates lower mitochondrial oxygen consumption, myofiber contractility, and exercise endurance. Respirometry analyses show that in vitro complex I respiration is unaffected by loss of SDH subunit C in muscle mitochondria, which is consistent with the pulmonary emphysema animal data. SDH knockout initially causes succinate accumulation associated with a down-regulated transcriptome but modest proteome effects. Muscle mass, myofiber type composition, and overall body mass constituents remain unaltered in the transgenic mice. Thus, while SDH regulates myofiber respiration in experimental pulmonary emphysema, it does not control muscle mass or other body constituents.


Subject(s)
Cell Respiration , Mice, Knockout , Muscle Contraction , Muscle, Skeletal , Pulmonary Emphysema , Succinate Dehydrogenase , Animals , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/genetics , Pulmonary Emphysema/pathology , Pulmonary Emphysema/etiology , Succinate Dehydrogenase/metabolism , Succinate Dehydrogenase/genetics , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Electron Transport Complex II/metabolism , Electron Transport Complex II/genetics , Disease Models, Animal , Mice, Transgenic , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/pathology , Oxygen Consumption
2.
PLoS One ; 19(7): e0305911, 2024.
Article in English | MEDLINE | ID: mdl-39052574

ABSTRACT

Chronic obstructive pulmonary disease (COPD), an inflammatory lung disease, causes approximately 3 million deaths each year; however, its pathological mechanisms are not fully understood. In this study, we examined whether HX110B, a mixture of Taraxacum officinale, Dioscorea batatas, and Schizonepeta tenuifolia extracts, could suppress porcine pancreatic elastase (PPE)-induced emphysema in mice and its mechanism of action. The therapeutic efficacy of HX110B was tested using a PPE-induced emphysema mouse model and human bronchial epithelial cell line BEAS-2B. In vivo data showed that the alveolar wall and air space expansion damaged by PPE were improved by HX110B administration. HX110B also effectively suppresses the expression levels of pro-inflammatory mediators including IL-6, IL-1ß, MIP-2, and iNOS, while stimulating the expression of lung protective factors such as IL-10, CC16, SP-D, and sRAGE. Moreover, HX110B improved the impaired OXPHOS subunit gene expression. In vitro analysis revealed that HX110B exerted its effects by activating the PPAR-RXR signaling pathways. Overall, our data demonstrated that HX110B could be a promising therapeutic option for COPD treatment.


Subject(s)
Pancreatic Elastase , Plant Extracts , Signal Transduction , Animals , Signal Transduction/drug effects , Mice , Pancreatic Elastase/metabolism , Humans , Plant Extracts/pharmacology , Pulmonary Emphysema/drug therapy , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/chemically induced , Pulmonary Emphysema/pathology , Peroxisome Proliferator-Activated Receptors/metabolism , Disease Models, Animal , Cell Line , Male , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Mice, Inbred C57BL , Swine
3.
Tissue Cell ; 89: 102452, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38986345

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is a prevalent and preventable condition. Mesenchymal stem cell (MSC) therapy is being explored to aid in the regeneration of lung cells and airway structure, aiming to restore lung function. AIM: To examine varied responses of MSCs when cultured with peripheral blood mononuclear cells (PBMCs) from different COPD phenotypes, patients were grouped into ACOS, emphysema, and chronic bronchitis categories. METHODS: PBMCs from these groups and controls were co-cultured with MSCs derived from dental follicles, revealing differing rates of apoptosis among COPD phenotypes compared to controls. RESULTS: While the chronic bronchitis group exhibited the least lymphocyte viability (p<0.01), introducing MSCs notably enhanced viability across all phenotypes except emphysema, with the chronic bronchitis group showing the most improvement (p<0.05). CONCLUSION: Stem cell therapy might reduce peripheral lymphocyte apoptosis in COPD, with varying responses based on phenotype, necessitating further research to understand mechanisms and optimize tailored therapies for each COPD subtype.


Subject(s)
Apoptosis , Bronchitis, Chronic , Pulmonary Disease, Chronic Obstructive , T-Lymphocytes , Humans , Pulmonary Disease, Chronic Obstructive/therapy , Pulmonary Disease, Chronic Obstructive/pathology , Male , Bronchitis, Chronic/therapy , Bronchitis, Chronic/pathology , Female , Middle Aged , T-Lymphocytes/immunology , Aged , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cell Transplantation , Pulmonary Emphysema/therapy , Pulmonary Emphysema/pathology , Emphysema/therapy , Emphysema/pathology
4.
Aging (Albany NY) ; 16(13): 10670-10693, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38954512

ABSTRACT

Senescent cells contribute to tissue aging and underlie the pathology of chronic diseases. The benefits of eliminating senescent cells have been demonstrated in several disease models, and the efficacy of senolytic drugs is currently being tested in humans. Exercise training has been shown to reduce cellular senescence in several tissues; however, the mechanisms responsible remain unclear. We found that myocyte-derived factors significantly extended the replicative lifespan of fibroblasts, suggesting that myokines mediate the anti-senescence effects of exercise. A number of proteins within myocyte-derived factors were identified by mass spectrometry. Among these, pigment epithelium-derived factor (PEDF) exerted inhibitory effects on cellular senescence. Eight weeks of voluntary running increased Pedf levels in skeletal muscles and suppressed senescence markers in the lungs. The administration of PEDF reduced senescence markers in multiple tissues and attenuated the decline in respiratory function in the pulmonary emphysema mouse model. We also showed that blood levels of PEDF inversely correlated with the severity of COPD in patients. Collectively, these results strongly suggest that PEDF contributes to the beneficial effects of exercise, potentially suppressing cellular senescence and its associated pathologies.


Subject(s)
Cellular Senescence , Eye Proteins , Lung , Nerve Growth Factors , Physical Conditioning, Animal , Serpins , Serpins/metabolism , Nerve Growth Factors/metabolism , Animals , Eye Proteins/metabolism , Mice , Lung/metabolism , Lung/pathology , Humans , Physical Conditioning, Animal/physiology , Male , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Fibroblasts/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Female , Muscle, Skeletal/metabolism , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology
5.
Int Immunopharmacol ; 139: 112680, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39018689

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a common disease with high global morbidity and mortality. Macrophages release IL-1ß and orchestrate airway inflammation in COPD. Previously, we explored the role of a new lncRNA, LincR-PPP2R5C, in regulating Th2 cells in asthma. Here, we established a murine model of COPD and explored the roles and mechanisms by which LincR-PPP2R5C regulates IL-1ß in macrophages. LincR-PPP2R5C was highly expressed in pulmonary macrophages from COPD-like mice. LincR-PPP2R5C deficiency ameliorated emphysema and pulmonary inflammation, as characterized by reduced IL-1ß in macrophages. Unexpectedly, in both lung tissues and macrophages, LincR-PPP2R5C deficiency decreased the expression of the IL-1ß protein but not the IL-1ß mRNA. Furthermore, we found that LincR-PPP2R5C deficiency increased the level of ubiquitinated IL-1ß in macrophages, which was mediated by PP2A activity. Targeting PP2A with FTY720 decreased IL-1ß and improved COPD. In conclusion, LincR-PPP2R5C regulates IL-1ß ubiquitination by affecting PP2A activity in macrophages, contributing to the airway inflammation and emphysema in a murine model of COPD. PP2A and IL-1ß ubiquitination in macrophages might be new therapeutic avenues for COPD therapy.


Subject(s)
Disease Models, Animal , Interleukin-1beta , Mice, Inbred C57BL , Pulmonary Disease, Chronic Obstructive , RNA, Long Noncoding , Ubiquitination , Animals , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/metabolism , Interleukin-1beta/metabolism , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Protein Phosphatase 2/metabolism , Macrophages/immunology , Macrophages/metabolism , Humans , Male , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/immunology , Pulmonary Emphysema/pathology , Pulmonary Emphysema/genetics , Lung/pathology , Lung/immunology , Mice, Knockout
6.
J Immunol ; 213(1): 75-85, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38758115

ABSTRACT

In chronic obstructive pulmonary disease (COPD), inflammation gives rise to protease-mediated degradation of the key extracellular matrix protein, elastin, which causes irreversible loss of pulmonary function. Intervention against proteolysis has met with limited success in COPD, due in part to our incomplete understanding of the mechanisms that underlie disease pathogenesis. Peptidyl arginine deiminase (PAD) enzymes are a known modifier of proteolytic susceptibility, but their involvement in COPD in the lungs of affected individuals is underexplored. In this study, we showed that enzyme isotypes PAD2 and PAD4 are present in primary granules of neutrophils and that cells from people with COPD release increased levels of PADs when compared with neutrophils of healthy control subjects. By examining bronchoalveolar lavage and lung tissue samples of patients with COPD or matched smoking and nonsmoking counterparts with normal lung function, we reveal that COPD presents with markedly increased airway concentrations of PADs. Ex vivo, we established citrullinated elastin in the peripheral airways of people with COPD, and in vitro, elastin citrullination significantly enhanced its proteolytic degradation by serine and matrix metalloproteinases, including neutrophil elastase and matrix metalloprotease-12, respectively. These results provide a mechanism by which neutrophil-released PADs affect lung function decline, indicating promise for the future development of PAD-based therapeutics for preserving lung function in patients with COPD.


Subject(s)
Elastin , Neutrophils , Protein-Arginine Deiminase Type 2 , Protein-Arginine Deiminase Type 4 , Proteolysis , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Humans , Neutrophils/immunology , Elastin/metabolism , Female , Male , Protein-Arginine Deiminase Type 4/metabolism , Middle Aged , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Pulmonary Emphysema/immunology , Aged , Protein-Arginine Deiminase Type 2/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Citrullination , Protein-Arginine Deiminases/metabolism , Leukocyte Elastase/metabolism , Lung/immunology , Lung/pathology
7.
Ann Surg Oncol ; 31(8): 5055-5063, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38739235

ABSTRACT

BACKGROUND: Emphysema is generally considered a poor prognostic factor for patients with nonsmall cell lung cancer; however, whether the poor prognosis is due to highly malignant tumors or emphysema itself remains unclear. This study was designed to determine the prognostic value of emphysema in patients with early-stage nonsmall cell lung cancer. METHODS: A total of 721 patients with clinical stage IA nonsmall cell lung cancer who underwent complete resection between April 2007 and December 2018 were retrospectively analyzed regarding clinicopathological findings and prognosis related to emphysema. RESULTS: The emphysematous and normal lung groups comprised 197 and 524 patients, respectively. Compared with the normal lung group, lymphatic invasion (23.9% vs. 14.1%, P = 0.003), vascular invasion (37.6% vs. 17.2%, P < 0.001), and pleural invasion (18.8% vs. 10.9%, P = 0.006) were observed more frequently in the emphysema group. Additionally, the 5-year overall survival rate was lower (77.1% vs. 91.4%, P < 0.001), and the cumulative incidence of other causes of death was higher in the emphysema group (14.0% vs. 3.50%, P < 0.001). Multivariable Cox regression analysis of overall survival revealed that emphysema (vs. normal lung, hazard ratio 2.02, P = 0.0052), age > 70 years (vs. < 70 years, hazard ratio 4.03, P < 0.001), and SUVmax > 1.8 (vs. ≤ 1.8, hazard ratio 2.20, P = 0.0043) were independent prognostic factors. CONCLUSIONS: Early-stage nonsmall cell lung cancer with emphysema has a tendency for the development of highly malignant tumors. Additionally, emphysema itself may have an impact on poor prognosis.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Pneumonectomy , Pulmonary Emphysema , Humans , Carcinoma, Non-Small-Cell Lung/surgery , Carcinoma, Non-Small-Cell Lung/pathology , Male , Lung Neoplasms/surgery , Lung Neoplasms/pathology , Lung Neoplasms/mortality , Female , Retrospective Studies , Survival Rate , Prognosis , Middle Aged , Aged , Follow-Up Studies , Pulmonary Emphysema/surgery , Pulmonary Emphysema/pathology , Pulmonary Emphysema/complications , Neoplasm Staging , Emphysema/surgery , Emphysema/pathology , Emphysema/etiology , Neoplasm Invasiveness
8.
Elife ; 132024 May 09.
Article in English | MEDLINE | ID: mdl-38722677

ABSTRACT

Environmental air irritants including nanosized carbon black (nCB) can drive systemic inflammation, promoting chronic obstructive pulmonary disease (COPD) and emphysema development. The let-7 microRNA (Mirlet7 miRNA) family is associated with IL-17-driven T cell inflammation, a canonical signature of lung inflammation. Recent evidence suggests the Mirlet7 family is downregulated in patients with COPD, however, whether this repression conveys a functional consequence on emphysema pathology has not been elucidated. Here, we show that overall expression of the Mirlet7 clusters, Mirlet7b/Mirlet7c2 and Mirlet7a1/Mirlet7f1/Mirlet7d, are reduced in the lungs and T cells of smokers with emphysema as well as in mice with cigarette smoke (CS)- or nCB-elicited emphysema. We demonstrate that loss of the Mirlet7b/Mirlet7c2 cluster in T cells predisposed mice to exaggerated CS- or nCB-elicited emphysema. Furthermore, ablation of the Mirlet7b/Mirlet7c2 cluster enhanced CD8+IL17a+ T cells (Tc17) formation in emphysema development in mice. Additionally, transgenic mice overexpressing Mirlet7g in T cells are resistant to Tc17 and CD4+IL17a+ T cells (Th17) development when exposed to nCB. Mechanistically, our findings reveal the master regulator of Tc17/Th17 differentiation, RAR-related orphan receptor gamma t (RORγt), as a direct target of Mirlet7 in T cells. Overall, our findings shed light on the Mirlet7/RORγt axis with Mirlet7 acting as a molecular brake in the generation of Tc17 cells and suggest a novel therapeutic approach for tempering the augmented IL-17-mediated response in emphysema.


Subject(s)
Cell Differentiation , Down-Regulation , MicroRNAs , Nuclear Receptor Subfamily 1, Group F, Member 3 , Pulmonary Emphysema , Th17 Cells , Animals , Female , Humans , Male , Mice , Interleukin-17/metabolism , Interleukin-17/genetics , Lung/pathology , Lung/metabolism , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Pulmonary Emphysema/genetics , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Th17 Cells/immunology , Th17 Cells/metabolism
9.
Acta Biomater ; 181: 282-296, 2024 06.
Article in English | MEDLINE | ID: mdl-38705223

ABSTRACT

Irreversible alveolar airspace enlargement is the main characteristic of pulmonary emphysema, which has been extensively studied using animal models. While the alterations in lung mechanics associated with these morphological changes have been documented in the literature, the study of the mechanical behavior of parenchymal tissue from emphysematous lungs has been poorly investigated. In this work, we characterize the mechanical and morphological properties of lung tissue in elastase-induced emphysema rat models under varying severity conditions. We analyze the non-linear tissue behavior using suitable hyperelastic constitutive models that enable to compare different non-linear responses in terms of hyperelastic material parameters. We further analyze the effect of the elastase dose on alveolar morphology and tissue material parameters and study their connection with respiratory-system mechanical parameters. Our results show that while the lung mechanical function is not significantly influenced by the elastase treatment, the tissue mechanical behavior and alveolar morphology are markedly affected by it. We further show a strong association between alveolar enlargement and tissue softening, not evidenced by respiratory-system compliance. Our findings highlight the importance of understanding tissue mechanics in emphysematous lungs, as changes in tissue properties could detect the early stages of emphysema remodeling. STATEMENT OF SIGNIFICANCE: Gas exchange is vital for life and strongly relies on the mechanical function of the lungs. Pulmonary emphysema is a prevalent respiratory disease where alveolar walls are damaged, causing alveolar enlargement that induces harmful changes in the mechanical response of the lungs. In this work, we study how the mechanical properties of lung tissue change during emphysema. Our results from animal models show that tissue properties are more sensitive to alveolar enlargement due to emphysema than other mechanical properties that describe the function of the whole respiratory system.


Subject(s)
Pancreatic Elastase , Pulmonary Emphysema , Animals , Pulmonary Emphysema/pathology , Pulmonary Emphysema/physiopathology , Lung/pathology , Rats , Male , Pulmonary Alveoli/pathology , Biomechanical Phenomena
10.
Immun Inflamm Dis ; 12(4): e1252, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38652015

ABSTRACT

We developed pulmonary emphysema and a type 2 airway inflammation overlap mouse model. The bronchoalveolar lavage (BAL) interleukin 13 (IL-13), IL-4, and IL-5 levels in the overlap model were higher than in the pulmonary emphysema model and lower than in the type 2 airway inflammation model, but IL-33 level in the lung was higher than in other models. IL-33 and interferon-γ (IFNγ) in lungs may control the severity of a type 2 airway inflammation in lung.


Subject(s)
Disease Models, Animal , Interleukin-33 , Pulmonary Emphysema , Animals , Interleukin-33/metabolism , Mice , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Pulmonary Emphysema/etiology , Pulmonary Emphysema/immunology , Bronchoalveolar Lavage Fluid/immunology , Lung/pathology , Lung/immunology , Lung/metabolism , Inflammation/immunology , Inflammation/metabolism , Interferon-gamma/metabolism , Interferon-gamma/immunology , Mice, Inbred C57BL
11.
J Ethnopharmacol ; 332: 118216, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-38642622

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Medicinal plants have shown promise in the search for new treatments of pulmonary emphysema. Anadenanthera colubrina, a species native to the Caatinga biome in northeastern Brazil, is widely recognized and traditionally employed in the treatment of pulmonary diseases. Many studies corroborate popular knowledge about the medicinal applications of A. colubrina, which has demonstrated a remarkable variety of pharmacological properties, however, its anti-inflammatory and antioxidant properties are highlighted. AIM OF THE STUDY: The objective of this study was to investigate the anti-inflammatory potential of the crude hydroethanolic extract of A. colubrina var. cebil (Griseb.) Altschul on pulmonary emphysema in rats as well as to determine its potential genotoxic and cytotoxic effects using the micronucleus assay. MATERIALS AND METHODS: The stem bark of the plant was collected in Pimenteiras-PI and sample was extracted by maceration using 70% ethanol. A portion of the extract underwent phytochemical analyses using TLC and HPLC. In this study, 8-week-old, male Wistar rats weighing approximately ±200 g was utilized following approval by local ethics committee for animal experimentation (No. 718/2022). Pulmonary emphysema was induced through orotracheal instillation of elastase, and treatment with A. colubrina extract or dexamethasone (positive control) concomitantly during induction. Twenty-eight days after the initiation of the protocol, plasma was used for cytokine measurement. Bronchoalveolar lavage (BAL) was used for leukocyte count. After euthanasia, lung samples were processed for histological analysis and quantification of oxidative stress markers. The micronucleus test was performed by evaluating the number of polychromatic erythrocytes (PCE) with micronuclei (MNPCE) to verify potential genotoxic effects of A. colubrina. A differential count of PCE and normochromatic erythrocytes (NCE) was performed to verify the potential cytotoxicity of the extract. Parametric data were subjected to normality analysis and subsequently to analysis of variance and Tukey or Dunnett post-test, non-parametric data were treated using the Kruskal-Wallis test with Dunn's post-test for unpaired samples. P value < 0.05 were considered significant. RESULTS: The A. colubrina extract did not show a significant increase in the number of MNPCE (p > 0.05), demonstrating low genotoxicity. No changes were observed in the PCE/NCE ratio of treated animals, compared with the vehicle, suggesting low cytotoxic potential of the extract. A significant reduction (p < 0.05) in neutrophilic inflammation was observed in the lungs of rats treated with the extract, evidenced by presence of these cells in both the tissue and BAL. The extract also demonstrated pulmonary antioxidant activity, with a significant decrease (p < 0.05) in myeloperoxidase, malondialdehyde, and nitrite levels. TNFα, IL-1ß, and IL-6 levels, as well as alveolar damage, were significantly reduced in animals treated with A. colubrina extract. Phytochemical analyses identified the presence of phenolic compounds and hydrolysable tannins in the A. colubrina extract. CONCLUSIONS: The findings of this study highlights the safety of the hydroethanolic extract of Anadenanthera colubrina, and demonstrates its potential as a therapeutic approach in the treatment of emphysema. The observed properties of this medicinal plant provide an optimistic outlook in the development of therapies for the treatment of pulmonary emphysema.


Subject(s)
Anti-Inflammatory Agents , Pancreatic Elastase , Plant Extracts , Pulmonary Emphysema , Rats, Wistar , Animals , Plant Extracts/pharmacology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/isolation & purification , Male , Pulmonary Emphysema/drug therapy , Pulmonary Emphysema/chemically induced , Pulmonary Emphysema/pathology , Rats , Plant Bark/chemistry , Disease Models, Animal , Bronchoalveolar Lavage Fluid/cytology , Lung/drug effects , Lung/pathology , Lung/metabolism , Micronucleus Tests , Oxidative Stress/drug effects
12.
Front Immunol ; 15: 1325090, 2024.
Article in English | MEDLINE | ID: mdl-38348034

ABSTRACT

Smoking is a leading risk factor of chronic obstructive pulmonary disease (COPD), that is characterized by chronic lung inflammation, tissue remodeling and emphysema. Although inflammation is critical to COPD pathogenesis, the cellular and molecular basis underlying smoking-induced lung inflammation and pathology remains unclear. Using murine smoke models and single-cell RNA-sequencing, we show that smoking establishes a self-amplifying inflammatory loop characterized by an influx of molecularly heterogeneous neutrophil subsets and excessive recruitment of monocyte-derived alveolar macrophages (MoAM). In contrast to tissue-resident AM, MoAM are absent in homeostasis and characterized by a pro-inflammatory gene signature. Moreover, MoAM represent 46% of AM in emphysematous mice and express markers causally linked to emphysema. We also demonstrate the presence of pro-inflammatory and tissue remodeling associated MoAM orthologs in humans that are significantly increased in emphysematous COPD patients. Inhibition of the IRAK4 kinase depletes a rare inflammatory neutrophil subset, diminishes MoAM recruitment, and alleviates inflammation in the lung of cigarette smoke-exposed mice. This study extends our understanding of the molecular signaling circuits and cellular dynamics in smoking-induced lung inflammation and pathology, highlights the functional consequence of monocyte and neutrophil recruitment, identifies MoAM as key drivers of the inflammatory process, and supports their contribution to pathological tissue remodeling.


Subject(s)
Emphysema , Pneumonia , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Humans , Mice , Animals , Macrophages, Alveolar/pathology , Monocytes/pathology , Pneumonia/pathology , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Emphysema/etiology , Pulmonary Emphysema/pathology , Inflammation/pathology , Emphysema/pathology
13.
Transl Res ; 269: 76-93, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38325750

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a prevalent lung disease usually resulting from cigarette smoking (CS). Cigarette smoking induces oxidative stress, which causes inflammation and alveolar epithelial cell apoptosis and represents a compelling therapeutic target for COPD. Purified human platelet-derived exosome product (PEP) is endowed with antioxidant enzymes and immunomodulatory molecules that mediate tissue repair. In this study, a murine model of CS-induced emphysema was used to determine whether nebulized PEP can influence the development of CS-induced emphysema through the mitigation of oxidative stress and inflammation in the lung. Nebulization of PEP effectively delivered the PEP vesicles into the alveolar region, with evidence of their uptake by type I and type II alveolar epithelial cells and macrophages. Lung function testing and morphometric assessment showed a significant attenuation of CS-induced emphysema in mice treated with nebulized PEP thrice weekly for 4 weeks. Whole lung immuno-oncology RNA sequencing analysis revealed that PEP suppressed several CS-induced cell injuries and inflammatory pathways. Validation of inflammatory cytokines and apoptotic protein expression on the lung tissue revealed that mice treated with PEP had significantly lower levels of S100A8/A9 expressing macrophages, higher levels of CD4+/FOXP3+ Treg cells, and reduced NF-κB activation, inflammatory cytokine production, and apoptotic proteins expression. Further validation using in vitro cell culture showed that pretreatment of alveolar epithelial cells with PEP significantly attenuated CS extract-induced apoptotic cell death. These data show that nebulization of exosomes like PEP can effectively deliver exosome cargo into the lung, mitigate CS-induced emphysema in mice, and suppress oxidative lung injury, inflammation, and apoptotic alveolar epithelial cell death.


Subject(s)
Blood Platelets , Cigarette Smoking , Extracellular Vesicles , Mice, Inbred C57BL , Pulmonary Emphysema , Animals , Extracellular Vesicles/metabolism , Pulmonary Emphysema/pathology , Pulmonary Emphysema/etiology , Mice , Cigarette Smoking/adverse effects , Blood Platelets/metabolism , Humans , Nebulizers and Vaporizers , Oxidative Stress/drug effects , Male , Apoptosis/drug effects
14.
Tohoku J Exp Med ; 263(1): 51-54, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38355110

ABSTRACT

Indium lung is an occupational lung disease caused by exposure to indium-tin-oxide (ITO) dust. Compared to other occupational lung diseases, indium lung has a shorter latency period and the respiratory status continues to worsen even after exposure to the work environment improves. Paraseptal emphysema which affects mainly the subpleural area is seen on chest images obtained via computed tomography (CT), regardless of the smoking history. However, the pathogenesis of emphysema in indium lung is still unclear. Therefore, we re-evaluated the pathology of three previously reported cases of indium lung. Paraseptal emphysema was observed in both smokers and nonsmokers. Obstructive respiratory impairment worsened over time in the cases with paraseptal emphysema. Many alveolar walls were destroyed independent of the presence or absence of emphysetamous changes or fibrosis. Moreover, bronchiolitis was found to be less common in indium lung than in asbestosis (the most common occupational lung disease) or common cases of chronic obstructive pulmonary disease caused by smoking. It has been shown that ITO causes protease anti-protease imbalance, oxidant-antioxidant imbalance, and continuous, abnormal inflammation (the three major causes of emphysema). In addition, nano-sized ITO is less likely to be trapped in the upper airways and may easily reach the subpleural alveoli. Furthermore, ITO may continue to cause sustained tissue injury at the alveolar level potentially resulting in emphysema. Further studies are needed to elucidate the detailed pathogenesis of indium lung by comparing it with other occupational lung diseases.


Subject(s)
Indium , Lung , Pulmonary Emphysema , Humans , Indium/toxicity , Lung/pathology , Lung/diagnostic imaging , Occupational Exposure/adverse effects , Pulmonary Emphysema/pathology , Pulmonary Emphysema/diagnostic imaging , Tin Compounds , Tomography, X-Ray Computed
15.
Am J Respir Cell Mol Biol ; 70(6): 482-492, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38377392

ABSTRACT

Cigarette smoking is known to be the leading cause of chronic obstructive pulmonary disease (COPD). However, the detailed mechanisms have not been elucidated. PAF (platelet-activating factor), a potent inflammatory mediator, is involved in the pathogenesis of various respiratory diseases such as bronchial asthma and COPD. We focused on LPLAT9 (lysophospholipid acyltransferase 9), a biosynthetic enzyme of PAF, in the pathogenesis of COPD. LPLAT9 gene expression was observed in excised COPD lungs and single-cell RNA sequencing data of alveolar macrophages (AMs). LPLAT9 was predominant and upregulated in AMs, particularly monocyte-derived AMs, in patients with COPD. To identify the function of LPLAT9/PAF in AMs in the pathogenesis of COPD, we exposed systemic LPLAT9-knockout (LPALT9-/-) mice to cigarette smoke (CS). CS increased the number of AMs, especially the monocyte-derived fraction, which secreted MMP12 (matrix metalloprotease 12). Also, CS augmented LPLAT9 phosphorylation/activation on macrophages and, subsequently, PAF synthesis in the lung. The LPLAT9-/- mouse lung showed reduced PAF production after CS exposure. Intratracheal PAF administration accumulated AMs by increasing MCP1 (monocyte chemoattractant protein-1). After CS exposure, AM accumulation and subsequent pulmonary emphysema, a primary pathologic change of COPD, were reduced in LPALT9-/- mice compared with LPLAT9+/+ mice. Notably, these phenotypes were again worsened by LPLAT9+/+ bone marrow transplantation in LPALT9-/- mice. Thus, CS-induced LPLAT9 activation in monocyte-derived AMs aggravated pulmonary emphysema via PAF-induced further accumulation of AMs. These results suggest that PAF synthesized by LPLAT9 has an important role in the pathogenesis of COPD.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase , Macrophages, Alveolar , Mice, Knockout , Platelet Activating Factor , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Animals , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Humans , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Pulmonary Emphysema/genetics , Platelet Activating Factor/metabolism , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , Mice , Male , Mice, Inbred C57BL , Matrix Metalloproteinase 12/metabolism , Matrix Metalloproteinase 12/genetics , Lung/metabolism , Lung/pathology , Cigarette Smoking/adverse effects , Cigarette Smoking/metabolism , Female
16.
Histol Histopathol ; 39(7): 805-816, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38226432

ABSTRACT

The aim of this review is to update and synthesize the molecular mechanisms that lead to the heterogeneous effect on tissue remodeling observed in the two most important clinical phenotypes of chronic obstructive pulmonary disease (COPD), pulmonary emphysema (PE) and chronic bronchitis (CB). Clinical and experimental evidence suggests that this heterogeneous response to promote PE, CB, or both, is related to differentiated genetic, epigenetic, and molecular conditions. Specifically, a tendency toward PE could be related to a variant in the DSP gene, SIRT1 downregulation, macrophage polarization to M1, as well as the involvement of the noncanonical Wnt5A signaling pathway, among other alterations. Additionally, in advanced stages of COPD, PE development is potentiated by dysregulations in autophagy, which promotes senescence and subsequently cell apoptosis, through exacerbated inflammasome activation and release of caspases. On the other hand, CB or the pro-fibrotic phenotype could be potentiated by the downregulated activity of HDAC2, the activation of the TGF-ß/Smad or Wnt/ß-catenin signaling pathways, macrophage polarization to M2, upregulation of TIMP-1, and/or the presence of the epithelial-mesenchymal transition (EMT) mechanism. Interestingly, the upregulated activity of MMPs, especially MMP-9, is widely involved in the development of both phenotypes. Furthermore, MMP-9 and MMP-12 enhance the severity, perpetuation, and exacerbation of COPD, as well as the development of autoimmunity in this disease.


Subject(s)
Bronchitis, Chronic , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Humans , Pulmonary Emphysema/pathology , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/genetics , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/genetics , Bronchitis, Chronic/metabolism , Bronchitis, Chronic/pathology , Bronchitis, Chronic/genetics , Animals , Signal Transduction
17.
Surg Today ; 54(2): 113-121, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37271799

ABSTRACT

PURPOSE: The present study evaluated the sex-specific susceptibility to the development of emphysema in patients with smoking histories who underwent lung cancer surgeries. METHODS: Lung cancer patients with smoking histories who underwent lung resection at the University of Tsukuba Hospital, Japan, were enrolled. Radiologic emphysematous changes were analyzed using three-dimensional computed tomography (3D-CT). The volume proportion of emphysematous lung per unit of smoking and the relationship between emphysematous change and clinicopathologic factors were evaluated. RESULTS: Radiologic emphysematous changes analyzed using 3D-CT per pack-year smoked, defined as the Smoking-Emphysema Index (SEI), were greater in females than males. The difference was more profound in adenocarcinoma patients than in non-adenocarcinoma patients (0.70 ± 2.30 vs. 0.21 ± 0.28, P = 0.037). CONCLUSION: Female lung cancer patients are more susceptible to smoking-induced emphysema than males. The SEI may be an effective indicator for evaluating smoking-induced emphysema.


Subject(s)
Emphysema , Lung Neoplasms , Pulmonary Emphysema , Male , Humans , Female , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/surgery , Lung Neoplasms/pathology , Lung/diagnostic imaging , Lung/pathology , Pulmonary Emphysema/diagnostic imaging , Pulmonary Emphysema/etiology , Pulmonary Emphysema/pathology , Emphysema/diagnostic imaging , Emphysema/etiology , Emphysema/pathology , Tomography, X-Ray Computed/methods , Smoking/adverse effects
18.
Am J Respir Crit Care Med ; 209(6): 683-692, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38055196

ABSTRACT

Rationale: Small airway disease is an important pathophysiological feature of chronic obstructive pulmonary disease (COPD). Recently, "pre-COPD" has been put forward as a potential precursor stage of COPD that is defined by abnormal spirometry findings or significant emphysema on computed tomography (CT) in the absence of airflow obstruction. Objective: To determine the degree and nature of (small) airway disease in pre-COPD using microCT in a cohort of explant lobes/lungs. Methods: We collected whole lungs/lung lobes from patients with emphysematous pre-COPD (n = 10); Global Initiative for Chronic Obstructive Lung Disease (GOLD) stage I (n = 6), II (n = 6), and III/IV (n = 7) COPD; and controls (n = 10), which were analyzed using CT and microCT. The degree of emphysema and the number and morphology of small airways were compared between groups, and further correlations were investigated with physiologic measures. Airway and parenchymal pathology was also validated with histopathology. Measurements and Main Results: The numbers of transitional bronchioles and terminal bronchioles per milliliter of lung were significantly lower in pre-COPD and GOLD stages I, II, and III/IV COPD compared with controls. In addition, the number of alveolar attachments of the transitional bronchioles and terminal bronchioles was also lower in pre-COPD and all COPD groups compared with controls. We did not find any differences between the pre-COPD and COPD groups in CT or microCT measures. The percentage of emphysema on CT showed the strongest correlation with the number of small airways in the COPD groups. Histopathology showed an increase in the mean chord length and a decrease in alveolar surface density in pre-COPD and all GOLD COPD stages compared with controls. Conclusions: Lungs of patients with emphysematous pre-COPD already show fewer small airways and airway remodeling even in the absence of physiologic airway obstruction.


Subject(s)
Asthma , Emphysema , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Humans , Cross-Sectional Studies , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/diagnostic imaging , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Emphysema/complications , Pulmonary Emphysema/diagnostic imaging , Pulmonary Emphysema/pathology , Lung , Asthma/pathology , X-Ray Microtomography
19.
Biochem Biophys Res Commun ; 694: 149419, 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38145597

ABSTRACT

BACKGROUND: Increasing evidence indicates that bioactive lipid mediators are involved in chronic obstructive pulmonary disease (COPD) pathogenesis. Recently, glycero-lysophospholipids, such as lysophosphatidic acid (LysoPA) and lysophosphatidylserine (LysoPS), have been recognized as significant inflammation-related lipid mediators. However, their association with COPD remains unclear. METHODS: We used an elastase-induced murine emphysema model to analyze the levels of lysophospholipids and diacyl-phospholipids in the lungs. Additionally, we assessed the expression of LysoPS-related genes and published data on smokers. RESULTS: In the early phase of an elastase-induced murine emphysema model, the levels of LysoPS and its precursor (phosphatidylserine [PS]) were significantly reduced, without significant modulations in other glycero-lysophospholipids. Additionally, there was an upregulation in the expression of lysoPS receptors, specifically GPR34, observed in the lungs of a cigarette smoke-exposed mouse model and the alveolar macrophages of human smokers. Elastase stimulation induces GPR34 expression in a human macrophage cell line in vitro. CONCLUSIONS: Elastase-induced lung emphysema affects the LysoPS/PS-GPR34 axis, and cigarette smoking or elastase upregulates GPR34 expression in alveolar macrophages. This novel association may serve as a potential pharmacological target for COPD treatment.


Subject(s)
Emphysema , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Mice , Humans , Animals , Pancreatic Elastase , Pulmonary Emphysema/chemically induced , Pulmonary Emphysema/pathology , Pulmonary Disease, Chronic Obstructive/pathology , Emphysema/chemically induced , Lysophospholipids/metabolism
20.
Eur J Med Res ; 28(1): 460, 2023 Oct 25.
Article in English | MEDLINE | ID: mdl-37880804

ABSTRACT

BACKGROUND: Hypomethylation of the perforin gene promoter in CD4 + T cells, inflammation and oxidative stress, might be involved in alveolar septal cell apoptosis associated with emphysema in rats. This study aimed to investigate the effects of S-adenosylmethionine (SAM) on this kind of apoptosis in rats with autoimmune emphysema. METHODS: Twenty-four rats were randomly divided into three groups: a normal control group, a model group, and a SAM group. Pathological changes in lung tissues were observed, and the mean linear intercept (MLI) and mean alveolar number (MAN) were measured. The levels of anti-endothelial cell antibodies (AECA) in serum, alveolar septal cell apoptosis, perforin gene promotor methylation in CD4 + T cells in the spleen, and the levels of cytokines, malondialdehyde (MDA), and glutathione (GSH) and the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) in bronchoalveolar lavage fluid (BALF) were investigated. RESULTS: The MLI, apoptosis index (AI) of alveolar septal cells, levels of AECA in serum, and levels of tumour necrosis factor-α (TNF-α), matrix metalloproteinase-9 (MMP-9) and MDA in BALF were increased, while the MAN, methylation levels, and the activities of GSH, SOD and GSH-Px in BALF were decreased in the model group compared with those in the normal control group and the SAM group (all P < 0.05). The levels of interleukin-8 (IL-8) in BALF were greater in the model group than in the normal control group (P < 0.05). CONCLUSIONS: SAM protects against alveolar septal cell apoptosis, airway inflammation and oxidative stress in rats with autoimmune emphysema possibly by partly reversing the hypomethylation of the perforin gene promoter in CD4 + T cells.


Subject(s)
Emphysema , Pulmonary Emphysema , Humans , Rats , Animals , S-Adenosylmethionine/pharmacology , Rats, Sprague-Dawley , Perforin/pharmacology , Pulmonary Emphysema/pathology , Lung/pathology , Emphysema/pathology , Apoptosis , Glutathione/pharmacology , Inflammation/pathology , Superoxide Dismutase
SELECTION OF CITATIONS
SEARCH DETAIL