1
|
Xiao X, Ding Z, Shi Y, Zhang Q. Causal Role of Immune Cells in Chronic Obstructive Pulmonary Disease: A Two-Sample Mendelian Randomization Study. COPD 2024; 21:2327352. [PMID: 38573027 DOI: 10.1080/15412555.2024.2327352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/27/2024] [Indexed: 04/05/2024]
Abstract
Accumulating evidence has highlighted the importance of immune cells in the pathogenesis of chronic obstructive pulmonary disease (COPD). However, the understanding of the causal association between immunity and COPD remains incomplete due to the existence of confounding variables. In this study, we employed a two-sample Mendelian randomization (MR) analysis, utilizing the genome-wide association study database, to investigate the causal association between 731 immune-cell signatures and the susceptibility to COPD from a host genetics perspective. To validate the consistency of our findings, we utilized MR analysis results of lung function data to assess directional concordance. Furthermore, we employed MR-Egger intercept tests, Cochrane's Q test, MR-PRESSO global test, and "leave-one-out" sensitivity analyses to evaluate the presence of horizontal pleiotropy, heterogeneity, and stability, respectively. Inverse variance weighting results showed that seven immune phenotypes were associated with the risk of COPD. Analyses of heterogeneity and pleiotropy analysis confirmed the reliability of MR results. These results highlight the interactions between the immune system and the lungs. Further investigations into their mechanisms are necessary and will contribute to inform targeted prevention strategies for COPD.
Collapse
Affiliation(s)
- Xinru Xiao
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Ziqi Ding
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Yujia Shi
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Qian Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou NO.2 People's Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| |
Collapse
|
2
|
Villoria GEM, Fischer RG, Tinoco EMB, Meyle J, Loos BG. Periodontal disease: A systemic condition. Periodontol 2000 2024. [PMID: 39494478 DOI: 10.1111/prd.12616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024]
Abstract
For decades, periodontitis has been considered to be a local inflammatory disease of the periodontal tissues in the oral cavity. Initially, associations of periodontitis with a multitude of noncommunicable diseases were each studied separately, and relationships were shown. The associations of periodontitis with morbidities, such as cardiovascular diseases, rheumatoid arthritis, diabetes mellitus, respiratory diseases, have been demonstrated. As most such studies were cross-sectional in nature, questions about causality cannot be univocally answered. And periodontitis as an independent risk factor for one systemic disease, becomes even more difficult to assess since recently periodontitis has also been associated with multimorbidity. Periodontitis and many systemic diseases share environmental, lifestyle and genetic risk factors, and share immunopathology. Moreover, suffering from one common noncommunicable disease may increase the susceptibility for another such chronic disease; the systemic effects of one condition may be one of various risk factors for another such disease. The overarching effect of any systemic disease is it causing a pro-inflammatory state in the individual; this has also been shown for periodontitis. Moreover, in periodontitis a prothrombotic state and elevated immunological activity have been shown. As such, when we consider periodontal disease as another systemic disease, it can affect the susceptibility and progression of other systemic diseases, and importantly, vice versa. And with this, it is not surprising that periodontitis is associated with a variety of other noncommunicable diseases. The medical definition of a systemic disease includes diseases that affect different organs and systems. Thus, the aim of this opinion paper is to propose that periodontitis should be considered a systemic disease in its own right and that it affects the individual's systemic condition and wellbeing. The dental and medical profession and researchers alike, should adapt this paradigm shift, advancing periodontal disease out of its isolated anatomical location into the total of chronic noncommunicable diseases, being for some conditions a comorbid disease and, vice versa, comorbidities can affect initiation and progression of periodontal disease.
Collapse
Affiliation(s)
- German E M Villoria
- Department of Periodontology, School of Dentistry, Rio de Janeiro State University, Rio de Janeiro, Brazil
- Department of Periodontology, School of Dentistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo G Fischer
- Department of Periodontology, School of Dentistry, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Eduardo M B Tinoco
- Department of Periodontology, School of Dentistry, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Joerg Meyle
- Dental School, University of Berne, Berne, Switzerland
| | - Bruno G Loos
- Department of Periodontology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Alves LHV, Ito JT, Almeida FM, Oliveira LM, Stelmach R, Tibério LFLC, Sato MN, Lopes FDTQS. Phenotypes of regulatory T cells in different stages of COPD. Int Immunopharmacol 2024; 140:112765. [PMID: 39083931 DOI: 10.1016/j.intimp.2024.112765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/27/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Previous studies have shown that failure to control inflammatory processes mediated by regulatory T (Treg) cells contributes to chronic obstructive pulmonary disease (COPD) development and progression. The activity of Treg cells depends on their phenotypic characteristics: resting Treg (rTreg, CD3+CD4+CD25+FOXP3+CD25++CD45RA+) and activated Treg (aTreg, CD3+CD4+CD25+FOXP3+CD25+++CD45RA-) cells exhibit immunosuppressive activity, while cytokine-secreting T cells (FrIII, CD3+CD4+CD25+FOXP3+CD25++CD45RA-) exhibit proinflammatory activity. Previous findings have shown an increased density of cytokine-secreting T cells in COPD patients experiencing exacerbation. However, the methods for evaluating COPD under stable conditions are lacking. AIM To evaluate Treg cell phenotypes in patients with different stages of COPD under stable conditions. METHODS Peripheral blood mononuclear cells (PBMCs) were isolated from non-obstructed smokers and ex-smokers (NOS group, n = 19) and COPD patients at different stages (COPD I-II group, n = 25; COPD III-IV group, n = 25). The phenotypic characteristics of Treg cells and Th17 cells and their respective intracellular cytokines were analyzed by flow cytometry. RESULTS Both obstructed groups showed an increase in the proportion of rTregs, while the COPD III-IV group showed additional increases in total Treg and Th17 cells and in IL-10+ cells. There was an increase in proinflammatory mediators (CD3+CD4+IL-17+ cells; CD3+CD4+RORγt+ cells) in the COPD I-II group. In contrast, the NOS group demonstrated high proportions of proinflammatory Treg cells and proinflammatory CD8+ T cells (CD3+CD8+IL-17+). CONCLUSION Despite the increase in both total Treg cells and the rTreg phenotype from the early stages of COPD, there was a decrease in cells expressing IL-10, suggesting a failure in controlling the inflammatory process. These events precede the progression of the inflammatory process mediated by Th17 cells.
Collapse
Affiliation(s)
- Luan H V Alves
- Laboratory of Experimental Therapeutics - (LIM20), Department of Clinical Medicine, School of Medicine, University of São Paulo, Brazil
| | - Juliana T Ito
- Laboratory of Experimental Therapeutics - (LIM20), Department of Clinical Medicine, School of Medicine, University of São Paulo, Brazil
| | - Francine M Almeida
- Laboratory of Experimental Therapeutics - (LIM20), Department of Clinical Medicine, School of Medicine, University of São Paulo, Brazil
| | - Luana M Oliveira
- Laboratory of Dermatology and Immunodeficiencies - LIM56, Department of Dermatology, Tropical Medicine Institute of São Paulo, School of Medicine, University of São Paulo, Brazil
| | - Rafael Stelmach
- Pulmonary Division, Heart Institute (InCor), Clinics Hospital, School of Medicine, University of São Paulo, Brazil
| | - Lolanda F L C Tibério
- Pulmonary Division, Heart Institute (InCor), Clinics Hospital, School of Medicine, University of São Paulo, Brazil
| | - Maria N Sato
- Laboratory of Dermatology and Immunodeficiencies - LIM56, Department of Dermatology, Tropical Medicine Institute of São Paulo, School of Medicine, University of São Paulo, Brazil
| | - Fernanda D T Q S Lopes
- Laboratory of Experimental Therapeutics - (LIM20), Department of Clinical Medicine, School of Medicine, University of São Paulo, Brazil; Thoracic Surgery Research Laboratory (LIM61). Division of Thoracic Surgery. Instituto do Coracao do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, SP, Brazil.
| |
Collapse
|
4
|
Wang F, Barrero CA. Multi-Omics Analysis Identified Drug Repurposing Targets for Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2024; 25:11106. [PMID: 39456887 PMCID: PMC11507528 DOI: 10.3390/ijms252011106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/04/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Despite recent advances in chronic obstructive pulmonary disease (COPD) research, few studies have identified the potential therapeutic targets systematically by integrating multiple-omics datasets. This project aimed to develop a systems biology pipeline to identify biologically relevant genes and potential therapeutic targets that could be exploited to discover novel COPD treatments via drug repurposing or de novo drug discovery. A computational method was implemented by integrating multi-omics COPD data from unpaired human samples of more than half a million subjects. The outcomes from genome, transcriptome, proteome, and metabolome COPD studies were included, followed by an in silico interactome and drug-target information analysis. The potential candidate genes were ranked by a distance-based network computational model. Ninety-two genes were identified as COPD signature genes based on their overall proximity to signature genes on all omics levels. They are genes encoding proteins involved in extracellular matrix structural constituent, collagen binding, protease binding, actin-binding proteins, and other functions. Among them, 70 signature genes were determined to be druggable targets. The in silico validation identified that the knockout or over-expression of SPP1, APOA1, CTSD, TIMP1, RXFP1, and SMAD3 genes may drive the cell transcriptomics to a status similar to or contrasting with COPD. While some genes identified in our pipeline have been previously associated with COPD pathology, others represent possible new targets for COPD therapy development. In conclusion, we have identified promising therapeutic targets for COPD. This hypothesis-generating pipeline was supported by unbiased information from available omics datasets and took into consideration disease relevance and development feasibility.
Collapse
Affiliation(s)
| | - Carlos A. Barrero
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA 19140, USA;
| |
Collapse
|
5
|
Murphy MP, Zieger M, Henry M, Meleady P, Mueller C, McElvaney NG, Reeves EP. Citrullination, a novel posttranslational modification of elastin, is involved in COPD pathogenesis. Am J Physiol Lung Cell Mol Physiol 2024; 327:L600-L606. [PMID: 39137524 DOI: 10.1152/ajplung.00185.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/15/2024] Open
Abstract
Elastin is an extracellular matrix protein (ECM) that supports elasticity of the lung, and in patients with chronic obstructive pulmonary disease (COPD) and emphysema, the structural changes that reduce the amount of elastic recoil, lead to loss of pulmonary function. We recently demonstrated that elastin is a target of peptidyl arginine deiminase (PAD) enzyme-induced citrullination, thereby leading to enhanced susceptibility of this ECM protein to proteolysis. This study aimed to investigate the impact of PAD activity in vivo and furthermore assessed whether pharmacological inhibition of PAD activity protects against pulmonary emphysema. Using a Serpina1a-e knockout mouse model, previously shown to develop inflammation-mediated emphysema, we validated the involvement of PADs in airway disease. In line with emphysema development, intratracheal administration of lipopolysaccharide in combination with PADs provoked significant airspace enlargement (P < 0.001) and diminished lung function, including loss of lung tissue elastance (P = 0.0217) and increases in lung volumes (P = 0.0463). Intraperitoneal treatment of mice with the PAD inhibitor, BB-Cl-amidine, prevented PAD/LPS-mediated lung function decline and emphysema and reduced levels of citrullinated airway elastin (P = 0.0199). These results provide evidence for the impact of PADs on lung function decline, indicating promising potential for the future development of PAD-based therapeutics for preserving lung function in patients with COPD.NEW & NOTEWORTHY This study provides evidence for the impact of peptidyl arginine deiminase (PAD) enzymes on lung function decline, indicating promising potential for the future development of PAD-based therapeutics for preserving lung function in patients with COPD.
Collapse
Affiliation(s)
- Mark P Murphy
- Department of Medicine, Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Marina Zieger
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States
- Department of Ophthalmology, Tufts Medical Center, Center for Translational Ocular Immunology, Boston, Massachusetts, United States
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Christian Mueller
- Genomic Medicine Unit, Sanofi, Waltham, Massachusetts, United States
| | - Noel G McElvaney
- Department of Medicine, Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Emer P Reeves
- Department of Anaesthesia and Critical Care Medicine, Pulmonary Clinical Science, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
6
|
Khanna V, Singh K. MicroRNAs as promising drug delivery target to ameliorate chronic obstructive pulmonary disease using nano-carriers: a comprehensive review. Mol Cell Biochem 2024:10.1007/s11010-024-05110-0. [PMID: 39254870 DOI: 10.1007/s11010-024-05110-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/27/2024] [Indexed: 09/11/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a deteriorating condition triggered by various factors, such as smoking, free radicals, and air pollution. This worsening disease is characterized by narrowing and thickening of airways, painful cough, and dyspnea. In COPD, numerous genes as well as microRNA (miRNA) play a significant role in the pathogenesis of the disease. Many in vivo and in vitro studies suggest that upregulation or suppression of certain miRNAs are effective treatment options for COPD. They have been proven to be more beneficial than the current symptomatic treatments, such as bronchodilators and corticosteroids. MiRNAs play a crucial role in immune cell development and regulate inflammatory responses in various tissues. MiRNA treatment thus allows for precision therapy with improved outcomes. Nanoparticle drug delivery systems such as polymeric nanoparticles, inorganic nanoparticles, dendrimers, polymeric micelles, and liposomes are an efficient method to ensure the biodistribution of the miRNAs to the target site. Identification of the right nanoparticle depending on the requirements and compatibility is essential for achieving maximum therapeutic effect. In this review, we offer a thorough comprehension of the pathology and genetics of COPD and the significance of miRNAs concerning various pathologies of the lung, as potential targets for treating the disease. The present review offers the latest insights into the nanoparticle drug delivery systems that can efficiently carry and deliver miRNA or antagomirs to the specific target site and hence help in effective management of COPD.
Collapse
Affiliation(s)
- Vamika Khanna
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, 400056, India
| | - Kavita Singh
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai, 400056, India.
| |
Collapse
|
7
|
Dilixiati N, Lian M, Hou Z, Song J, Yang J, Lin R, Wang J. Nomograms for Predicting High Hospitalization Costs and Prolonged Stay among Hospitalized Patients with pAECOPD. Can Respir J 2024; 2024:2639080. [PMID: 39280690 PMCID: PMC11398965 DOI: 10.1155/2024/2639080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 06/24/2024] [Accepted: 08/17/2024] [Indexed: 09/18/2024] Open
Abstract
This study aimed to develop nomograms to predict high hospitalization costs and prolonged stays in hospitalized acute exacerbations of chronic obstructive pulmonary disease (AECOPD) patients with community-acquired pneumonia (CAP), also known as pAECOPD. A total of 635 patients with pAECOPD were included in this observational study and divided into training and testing sets. Variables were initially screened using univariate analysis, and then further selected using a backward stepwise regression. Multivariable logistic regression was performed to establish nomograms. The predictive performance of the model was evaluated using the receiver operating characteristic (ROC) curve, area under the curve (AUC), calibration curve, and decision curve analysis (DCA) in both the training and testing sets. Finally, the logistic regression analysis showed that elevated white blood cell count (WBC>10 × 109 cells/l), hypoalbuminemia, pulmonary encephalopathy, respiratory failure, diabetes, and respiratory intensive care unit (RICU) admissions were risk factors for predicting high hospitalization costs in pAECOPD patients. The AUC value was 0.756 (95% CI: 0.699-0.812) in the training set and 0.792 (95% CI: 0.718-0.867) in the testing set. The calibration plot and DCA curve indicated the model had good predictive performance. Furthermore, decreased total protein, pulmonary encephalopathy, reflux esophagitis, and RICU admissions were risk factors for predicting prolonged stays in pAECOPD patients. The AUC value was 0.629 (95% CI: 0.575-0.682) in the training set and 0.620 (95% CI: 0.539-0.701) in the testing set. The calibration plot and DCA curve indicated the model had good predictive performance. We developed and validated two nomograms for predicting high hospitalization costs and prolonged stay, respectively, among hospitalized patients with pAECOPD. This trial is registered with ChiCTR2000039959.
Collapse
Affiliation(s)
- Nafeisa Dilixiati
- Department of Pulmonary and Critical Care MedicineBeijing Luhe HospitalCapital Medical University, Beijing, China
| | - Mengyu Lian
- Department of Pulmonary and Critical Care MedicineBeijing Luhe HospitalCapital Medical University, Beijing, China
| | - Ziliang Hou
- Department of Pulmonary and Critical Care MedicineBeijing Luhe HospitalCapital Medical University, Beijing, China
| | - Jie Song
- Department of Pulmonary and Critical Care MedicineBeijing Luhe HospitalCapital Medical University, Beijing, China
| | - Jingjing Yang
- Department of Pulmonary and Critical Care MedicineBeijing Luhe HospitalCapital Medical University, Beijing, China
| | - Ruiyan Lin
- Department of Pulmonary and Critical Care MedicineBeijing Luhe HospitalCapital Medical University, Beijing, China
| | - Jinxiang Wang
- Department of Pulmonary and Critical Care MedicineBeijing Luhe HospitalCapital Medical University, Beijing, China
| |
Collapse
|
8
|
Robertoni FSZ, Velosa APP, Oliveira LDM, de Almeida FM, da Silveira LKR, Queiroz ZADJ, Lobo TDM, Contini VE, Baldavira CM, Carrasco S, Fernezlian SDM, Sato MN, Capelozzi VL, Lopes FDTQDS, Teodoro WPR. Type V collagen-induced nasal tolerance prevents lung damage in an experimental model: new evidence of autoimmunity to collagen V in COPD. Front Immunol 2024; 15:1444622. [PMID: 39301030 PMCID: PMC11410637 DOI: 10.3389/fimmu.2024.1444622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/16/2024] [Indexed: 09/22/2024] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) has been linked to immune responses to lung-associated self-antigens. Exposure to cigarette smoke (CS), the main cause of COPD, causes chronic lung inflammation, resulting in pulmonary matrix (ECM) damage. This tissue breakdown exposes collagen V (Col V), an antigen typically hidden from the immune system, which could trigger an autoimmune response. Col V autoimmunity has been linked to several lung diseases, and the induction of immune tolerance can mitigate some of these diseases. Evidence suggests that autoimmunity to Col V might also occur in COPD; thus, immunotolerance to Col V could be a novel therapeutic approach. Objective The role of autoimmunity against collagen V in COPD development was investigated by analyzing the effects of Col V-induced tolerance on the inflammatory response and lung remodeling in a murine model of CS-induced COPD. Methods Male C57BL/6 mice were divided into three groups: one exposed to CS for four weeks, one previously tolerated for Col V and exposed to CS for four weeks, and one kept in clean air for the same period. Then, we proceeded with lung functional and structural evaluation, assessing inflammatory cells in bronchoalveolar lavage fluid (BALF) and inflammatory markers in the lung parenchyma, inflammatory cytokines in lung and spleen homogenates, and T-cell phenotyping in the spleen. Results CS exposure altered the structure of elastic and collagen fibers and increased the pro-inflammatory immune response, indicating the presence of COPD. Col V tolerance inhibited the onset of emphysema and prevented structural changes in lung ECM fibers by promoting an immunosuppressive microenvironment in the lung and inducing Treg cell differentiation. Conclusion Induction of nasal tolerance to Col V can prevent inflammatory responses and lung remodeling in experimental COPD, suggesting that autoimmunity to Col V plays a role in COPD development.
Collapse
Affiliation(s)
| | | | - Luana de Mendonça Oliveira
- Laboratory of Dermatology and Immunodeficiencies, Laboratório de Investigação Médica (LIM)-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| | - Francine Maria de Almeida
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics, Laboratório de Investigação Médica (LIM)-20, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Thays de Matos Lobo
- Division of Rheumatology, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Vitória Elias Contini
- Division of Rheumatology, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | - Solange Carrasco
- Division of Rheumatology, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | - Maria Notomi Sato
- Laboratory of Dermatology and Immunodeficiencies, Laboratório de Investigação Médica (LIM)-56, Department of Dermatology, Tropical Medicine Institute of São Paulo, University of São Paulo Medical School, São Paulo, Brazil
| | - Vera Luiza Capelozzi
- Department of Pathology, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | | |
Collapse
|
9
|
Fu C, Shi J, Hu Y, Luo J. Association Between Dietary Selenium Intake and Chronic Obstructive Pulmonary Disease: A Cross-Sectional Study from the National Health and Nutrition Examination Survey 1999-2018. Int J Chron Obstruct Pulmon Dis 2024; 19:1893-1903. [PMID: 39206145 PMCID: PMC11350224 DOI: 10.2147/copd.s473664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a widespread respiratory condition characterized by chronic inflammation. Selenium, an essential trace element, possesses anti-inflammatory and antioxidant properties. However, the diet is often complex, and the absence of one nutrient may indicate a concurrent deficiency in others. Therefore, inadequate dietary selenium intake may suggest deficiencies in other elements. Despite its potential benefits, there is a scarcity of evidence regarding the association between dietary selenium intake and COPD. Purpose This study aims to investigate the potential association between dietary selenium intake and COPD among American adults. Patients and Methods This cross-sectional study analyzed data from the National Health and Nutrition Examination Survey conducted in the United States from 1999 to 2018. Multivariate logistic regression, restricted cubic spline analyses, subgroup analysis, and sensitivity analysis were conducted to assess the correlation between dietary selenium intake and COPD. Results A total of 39,654 participants were included in the study. The adjusted odds ratio (OR) for COPD in the highest selenium intake group (T3, > 122.0 μg/day) was 0.80 (95% CI: 0.71-0.91, p < 0.001) compared to the lowest intake group (T1, < 81.6 μg/day). Dietary selenium intake exhibited a linear negative correlation with COPD. Among participants reporting selenium supplementation, a similar negative association persisted. Conclusion This study observed a negative correlation between dietary selenium intake and COPD among American adults, indicating a possible association between higher selenium intake and a lower risk of COPD.
Collapse
Affiliation(s)
- Chengfeng Fu
- Respiratory and Critical Care Medicine, the Second People’s Hospital of Banan District, Chongqing, 400054, People’s Republic of China
| | - Junwei Shi
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu, 210023, People’s Republic of China
| | - Ying Hu
- Respiratory and Critical Care Medicine, the Second People’s Hospital of Banan District, Chongqing, 400054, People’s Republic of China
| | - Jing Luo
- Respiratory and Critical Care Medicine, the Second People’s Hospital of Banan District, Chongqing, 400054, People’s Republic of China
| |
Collapse
|
10
|
Distefano A, Orlando L, Giallongo S, Tropea E, Spampinato M, Santisi A, Longhitano L, Parisi G, Leonardi S, Russo A, Caruso M, Di Rosa M, Tibullo D, Salamone M, Li Volti G, Barbagallo IA. Fish Oil Containing Pro-Resolving Mediators Enhances the Antioxidant System and Ameliorates LPS-Induced Inflammation in Human Bronchial Epithelial Cells. Pharmaceuticals (Basel) 2024; 17:1066. [PMID: 39204170 PMCID: PMC11360764 DOI: 10.3390/ph17081066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/01/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
Fish oil, renowned for its high content of omega-3 fatty acids, particularly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), has gained considerable attention for its potential health benefits. EPA and DHA exhibit anti-inflammatory effects by promoting the production of specialized pro-resolving mediators (SPMs), such as resolvins and protectins. Fish oil has been studied for its potential to reduce bronchial inflammation, a key feature of respiratory conditions like asthma and COPD. This study investigates the cellular mechanisms of fish oil in an in vitro model of lung inflammation using lipopolysaccharide (LPS) on a healthy human bronchial epithelium cell line. LPS exposure for 24 h reduced cell viability, elevated reactive oxygen species (ROS), depleted glutathione (GSH), and induced mitochondrial depolarization, indicating oxidative stress and inflammation. Fish oil administration significantly mitigated ROS production, prevented GSH depletion, and reduced mitochondrial depolarization. This was associated with the upregulation of the endogenous antioxidant system, evidenced by restored GSH levels and the increased gene expression of glutathione peroxidase (GPX), catalase (CAT), superoxide dismutase 1 (SOD1), and superoxide dismutase 2 (SOD2). Fish oil also suppressed IL-6 and IL-1β expression and increased anti-inflammatory cytokine IL-10 expression. Furthermore, fish oil upregulated the expression of pro-resolving mediator receptors, suggesting a role in inflammation resolution. These findings highlight the potential of fish oil supplementation as a preventive measure against pulmonary diseases characterized by unresolved inflammation such as lung inflammation.
Collapse
Affiliation(s)
- Alfio Distefano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.D.); (L.O.); (E.T.); (M.S.); (A.S.); (L.L.); (M.C.); (M.D.R.); (D.T.); (M.S.); (G.L.V.); (I.A.B.)
| | - Laura Orlando
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.D.); (L.O.); (E.T.); (M.S.); (A.S.); (L.L.); (M.C.); (M.D.R.); (D.T.); (M.S.); (G.L.V.); (I.A.B.)
| | - Sebastiano Giallongo
- Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy
| | - Emanuela Tropea
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.D.); (L.O.); (E.T.); (M.S.); (A.S.); (L.L.); (M.C.); (M.D.R.); (D.T.); (M.S.); (G.L.V.); (I.A.B.)
| | - Mariarita Spampinato
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.D.); (L.O.); (E.T.); (M.S.); (A.S.); (L.L.); (M.C.); (M.D.R.); (D.T.); (M.S.); (G.L.V.); (I.A.B.)
| | - Annalisa Santisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.D.); (L.O.); (E.T.); (M.S.); (A.S.); (L.L.); (M.C.); (M.D.R.); (D.T.); (M.S.); (G.L.V.); (I.A.B.)
| | - Lucia Longhitano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.D.); (L.O.); (E.T.); (M.S.); (A.S.); (L.L.); (M.C.); (M.D.R.); (D.T.); (M.S.); (G.L.V.); (I.A.B.)
| | - Giuseppe Parisi
- Pediatric Respiratory Unit, AOUP “G. Rodolico-San Marco”, University of Catania, 95123 Catania, Italy; (G.P.); (S.L.)
| | - Salvatore Leonardi
- Pediatric Respiratory Unit, AOUP “G. Rodolico-San Marco”, University of Catania, 95123 Catania, Italy; (G.P.); (S.L.)
| | - Arcangelo Russo
- Department of Medicine, Kore University of Enna, 94100 Enna, Italy;
| | - Massimo Caruso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.D.); (L.O.); (E.T.); (M.S.); (A.S.); (L.L.); (M.C.); (M.D.R.); (D.T.); (M.S.); (G.L.V.); (I.A.B.)
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.D.); (L.O.); (E.T.); (M.S.); (A.S.); (L.L.); (M.C.); (M.D.R.); (D.T.); (M.S.); (G.L.V.); (I.A.B.)
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.D.); (L.O.); (E.T.); (M.S.); (A.S.); (L.L.); (M.C.); (M.D.R.); (D.T.); (M.S.); (G.L.V.); (I.A.B.)
| | - Maurizio Salamone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.D.); (L.O.); (E.T.); (M.S.); (A.S.); (L.L.); (M.C.); (M.D.R.); (D.T.); (M.S.); (G.L.V.); (I.A.B.)
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.D.); (L.O.); (E.T.); (M.S.); (A.S.); (L.L.); (M.C.); (M.D.R.); (D.T.); (M.S.); (G.L.V.); (I.A.B.)
| | - Ignazio Alberto Barbagallo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (A.D.); (L.O.); (E.T.); (M.S.); (A.S.); (L.L.); (M.C.); (M.D.R.); (D.T.); (M.S.); (G.L.V.); (I.A.B.)
| |
Collapse
|
11
|
Xie B, Chen Q, Dai Z, Jiang C, Chen X. Progesterone (P4) ameliorates cigarette smoke-induced chronic obstructive pulmonary disease (COPD). Mol Med 2024; 30:123. [PMID: 39138434 PMCID: PMC11323532 DOI: 10.1186/s10020-024-00883-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 07/19/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory lung disease associated with high morbidity and mortality worldwide. Oxidative injury and mitochondrial dysfunction in the airway epithelium are major events in COPD progression. METHODS AND RESULTS The therapeutic effects of Progesterone (P4) were investigated in vivo and in vitro in this study. In vivo, in a cigarette smoke (CS) exposure-induced COPD mouse model, P4 treatment significantly ameliorated CS exposure-induced physiological and pathological characteristics, including inflammatory cell infiltration and oxidative injury, in a dose-dependent manner. The c-MYC/SIRT1/PGC-1α pathway is involved in the protective function of P4 against CS-induced COPD. In vitro, P4 co-treatment significantly ameliorated H2O2-induced oxidative injury and mitochondrial dysfunctions by promoting cell proliferation, increasing mitochondrial membrane potential, decreasing ROS levels and apoptosis, and increasing ATP content. Moreover, P4 co-treatment partially attenuated H2O2-caused inhibition in Nrf1, Tfam, Mfn1, PGR-B, c-MYC, SIRT1, and PGC-1α levels. In BEAS-2B and ASM cells, the c-MYC/SIRT1 axis regulated P4's protective effects against H2O2-induced oxidative injury and mitochondrial dysfunctions. CONCLUSION P4 activates the c-MYC/SIRT1 axis, ameliorating CS-induced COPD and protecting both airway epithelial cells and smooth muscle cells against H2O2-induced oxidative damage. PGC-1α and downstream mitochondrial signaling pathways might be involved.
Collapse
Affiliation(s)
- Bin Xie
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Departement of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiong Chen
- Departement of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ziyu Dai
- Departement of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Chen Jiang
- Departement of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xi Chen
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
12
|
Tessier B, Moine L, Peramo A, Tsapis N, Fattal E. Poly(malic acid)-budesonide nanoconjugates embedded in microparticles for lung administration. Drug Deliv Transl Res 2024; 14:2062-2078. [PMID: 38517568 DOI: 10.1007/s13346-024-01571-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 03/24/2024]
Abstract
To improve the therapeutic activity of inhaled glucocorticoids and reduce potential side effects, we designed a formulation combining the advantages of nanoparticles, which have an enhanced uptake by alveolar cells, allow targeted delivery and sustained drug release, as well as limited drug systemic passage, with those of microparticles, which display good alveolar deposition. Herein, a polymer-drug conjugate, poly(malic acid)-budesonide (PMAB), was first synthesized with either 11, 20, 33, or 43 mol% budesonide (drug:polymer from 1:8 to 3:4), the drug creating hydrophobic domains. The obtained conjugates self-assemble into nanoconjugates in water, yielding excellent drug loading of up to 73 wt%, with 80-100 nm diameters. In vitro assays showed that budesonide could be steadily released from the nanoconjugates, and the anti-inflammatory activity was preserved, as evidenced by reduced cytokine production in LPS-activated RAW 264.7 macrophages. Nanoconjugates were then embedded into microparticles through spray-drying with L-leucine, forming nano-embedded microparticles (NEMs). NEMs were produced with an aerodynamic diameter close to 1 µm and a density below 0.1 g.cm-3, indicative of a high alveolar deposition. NEMs spray-dried with the less hydrophobic nanoconjugates, PMAB 1:4, were readily dissolved in simulated lung fluid and were chosen for in vivo experiments to study pharmacokinetics in healthy rats. As it was released in vivo from NEMs, sustained distribution of budesonide was obtained for 48 h in lung tissue, cells, and lining fluid. With high loading rates, modulable release kinetics, and low cytotoxicity, these nanoconjugates delivered by NEMs are promising for the more efficient treatment of pulmonary inflammatory diseases.
Collapse
Affiliation(s)
- Barbara Tessier
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | - Laurence Moine
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | - Arnaud Peramo
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | - Nicolas Tsapis
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | - Elias Fattal
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France.
| |
Collapse
|
13
|
Jiang Q, Jiang Y, Ma Z, Huang J, Li Y. Nonlinear correlation and mediation effects between serum 25-hydroxyvitamin D levels and all-cause mortality in COPD patients. Front Nutr 2024; 11:1412606. [PMID: 38903612 PMCID: PMC11188383 DOI: 10.3389/fnut.2024.1412606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 05/16/2024] [Indexed: 06/22/2024] Open
Abstract
Background Numerous studies have shown that low levels of vitamin D are linked to a higher risk of inflammatory diseases and their progression. However, how vitamin D levels affect mortality in chronic obstructive pulmonary disease (COPD) patients is still unclear. Thus, this study aimed to explore the relationship between serum 25-hydroxyvitamin D [25(OH)D] levels and the risk of death from all causes in U.S. adults with COPD. Methods This study analyzed 1,876 adults with COPD from the National Health and Nutrition Examination Survey (2005-2018). Mortality data up to December 31, 2019, were obtained from the National Death Index (NDI) records. Participants were categorized into three groups according to their 25(OH)D levels: Q1 (<50.0 nmol/L) for deficiency; Q2 (50.0-74.9 nmol/L) for insufficiency; and Q3 (≥75.0 nmol/L) for adequacy. A weighted Cox regression model assessed the link between 25(OH)D levels and mortality. Kaplan-Meier survival curves, subgroup, and sensitivity analyses were conducted. Additionally, the relationship between 25(OH)D and the hazard ratio (HR) was detailed through restricted cubic spline analysis. Mediation analysis revealed how 25(OH)D mediates the relationship between Dietary Inflammatory Index and mortality. Results There were 395 all-cause deaths during the follow-up, resulting in a mortality rate of 21.06%. After adjusting for potential confounders, higher 25(OH)D levels significantly correlated with a lower risk of all-cause mortality in COPD patients (HR = 0.52, 95% CI: 0.37-0.72, p < 0.001). Restricted cubic spline analysis indicated a non-linear relationship between 25(OH)D levels and all-cause mortality (p for nonlinear = 0.023), with levels below 63.4 nmol/L posing an independent risk for all-cause mortality in COPD patients (HR = 0.98, 95% CI: 0.97-0.99, p = 0.005). Sensitivity and subgroup analyses confirmed our results' robustness, with mediation analysis showing 25(OH)D's 22% mediating effect on diet-induced inflammation and all-cause mortality in COPD patients. Conclusion 25(OH)D independently lowers the risk of all-cause mortality in COPD patients, with a non-linear L-shaped correlation, and mediates the effect of Dietary Inflammatory Index on mortality, suggesting new therapeutic possibilities.
Collapse
Affiliation(s)
- Qi Jiang
- Department of Respiratory and Critical Care Medicine, First Hospital of Jilin University, Changchun, China
| | - Yuewen Jiang
- Department of Respiratory and Critical Care Medicine, Qiyang People's Hospital, Yongzhou, China
| | - Zheru Ma
- Orthopaedic Center, First Hospital of Jilin University, Changchun, China
| | - Jingda Huang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Yang Li
- Department of Respiratory and Critical Care Medicine, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Zhang CL, Maccarone JR, Grady ST, Collins CM, Moy ML, Hart JE, Kang CM, Coull BA, Schwartz JD, Koutrakis P, Garshick E. Indoor and ambient black carbon and fine particulate matter associations with blood biomarkers in COPD patients. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 927:171897. [PMID: 38522542 PMCID: PMC11090036 DOI: 10.1016/j.scitotenv.2024.171897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Systemic inflammation contributes to cardiovascular risk and chronic obstructive pulmonary disease (COPD) pathophysiology. Associations between systemic inflammation and exposure to ambient fine particulate matter (PM ≤ 2.5 μm diameter; PM2.5), and black carbon (BC), a PM2.5 component attributable to traffic and other sources of combustion, infiltrating indoors are not well described. METHODS Between 2012 and 2017, COPD patients completed in-home air sampling over one-week intervals, up to four times (seasonally), followed by measurement of plasma biomarkers of systemic inflammation, C-reactive protein (CRP) and interleukin-6 (IL-6), and endothelial activation, soluble vascular adhesion molecule-1 (sVCAM-1). Ambient PM2.5, BC and sulfur were measured at a central site. The ratio of indoor/ambient sulfur in PM2.5, a surrogate for fine particle infiltration, was used to estimate indoor BC and PM2.5 of ambient origin. Linear mixed effects regression with a random intercept for each participant was used to assess associations between indoor and indoor of ambient origin PM2.5 and BC with each biomarker. RESULTS 144 participants resulting in 482 observations were included in the analysis. There were significant positive associations between indoor BC and indoor BC of ambient origin with CRP [%-increase per interquartile range (IQR);95 % CI (13.2 %;5.2-21.8 and 11.4 %;1.7-22.1, respectively)]. Associations with indoor PM2.5 and indoor PM2.5 of ambient origin were weaker. There were no associations with IL-6 or sVCAM-1. CONCLUSIONS In homes of patients with COPD without major sources of combustion, indoor BC is mainly attributable to the infiltration of ambient sources of combustion indoors. Indoor BC of ambient origin is associated with increases in systemic inflammation in patients with COPD, even when staying indoors.
Collapse
Affiliation(s)
- Cathy L Zhang
- Research and Development Service, VA Boston Healthcare System, 1400 VFW Parkway, West Roxbury, MA 02132, USA
| | - Jennifer R Maccarone
- Pulmonary, Allergy, Sleep, and Critical Care Medicine Section, Medical Service, VA Boston Healthcare System, 1400 VFW Parkway, West Roxbury, Boston, MA 02132, USA; The Pulmonary Center, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Stephanie T Grady
- Boston University School of Public Health, 715 Albany St, Boston, MA 02118, USA
| | - Christina M Collins
- Research and Development Service, VA Boston Healthcare System, 1400 VFW Parkway, West Roxbury, MA 02132, USA
| | - Marilyn L Moy
- Pulmonary, Allergy, Sleep, and Critical Care Medicine Section, Medical Service, VA Boston Healthcare System, 1400 VFW Parkway, West Roxbury, Boston, MA 02132, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Jaime E Hart
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Ave, Boston, MA 02115, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Choong-Min Kang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Ave, Boston, MA 02115, USA
| | - Brent A Coull
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Ave, Boston, MA 02115, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA
| | - Joel D Schwartz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Ave, Boston, MA 02115, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA
| | - Petros Koutrakis
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Ave, Boston, MA 02115, USA
| | - Eric Garshick
- Pulmonary, Allergy, Sleep, and Critical Care Medicine Section, Medical Service, VA Boston Healthcare System, 1400 VFW Parkway, West Roxbury, Boston, MA 02132, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA.
| |
Collapse
|
15
|
Yalcinsoy M, Beykumul A, Gulbas G, Arslan AK, Neselioglu S. DO disease stages affect oxidative stress in stable COPD? Heliyon 2024; 10:e31188. [PMID: 38831835 PMCID: PMC11145427 DOI: 10.1016/j.heliyon.2024.e31188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/09/2024] [Accepted: 05/12/2024] [Indexed: 06/05/2024] Open
Abstract
Background Detection of oxidative stress level may lead us to understand the pathogenesis of COPD better and to search for new treatments. Oxidative stress levels have also been shown to be elevated in stable COPD patients. We aimed to investigate whether the stage of COPD affects the severity of inflammation-induced oxidative stress in patients with stable COPD. Methods Between June 2019 and March 2020, all consecutive patients admitted to COPD-specific outpatient clinics were included. Patients were classified A, B, and E according to the GOLD guideline. Results The median age of 98 patients (Male: 92 (93.9 %)) was 65 (min-max: 49-86). A statistically significant difference was found between the groups in FEV1, FVC, and FEV1/FVC (p < 0.001). age, and thiols (r = -0.168, p = 0.049; r = -0.184, p = 0.035) and DS (r = -0.209, p = 0.019) were found to be negatively correlated at a low level. When adjusted for age, oxidative stress parameters were similar between stages. Conclusion No difference between stages and oxidative stress parameters according to GOLD classification in stable COPD patients. Our results may be a guide for not using anti-inflammatory therapy except for attacks.
Collapse
Affiliation(s)
- Murat Yalcinsoy
- Department of Pulmonary Medicine, Inonu University Medical Faculty, Turgut Ozal Medical Center, Malatya, Turkey
| | - Aysegul Beykumul
- Department of Pulmonary Rehabilitation, Inonu University Medical Faculty, Turgut Ozal Medical Center, Malatya, Turkey
| | - Gazi Gulbas
- Department of Pulmonary Rehabilitation, Inonu University Medical Faculty, Turgut Ozal Medical Center, Malatya, Turkey
| | | | - Salim Neselioglu
- Department of Biochemistry, Yıldırım Beyazıt University Medical Faculty, Ankara, Turkey
| |
Collapse
|
16
|
Aguirre-Franco C, Torres-Duque CA, Salazar G, Casas A, Jaramillo C, Gonzalez-Garcia M. Prevalence of pulmonary hypertension in COPD patients living at high altitude. Pulmonology 2024; 30:247-253. [PMID: 35151623 DOI: 10.1016/j.pulmoe.2021.12.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Pulmonary hypertension (PH) is associated with poor prognosis for patients with chronic obstructive pulmonary disease (COPD). Most of the knowledge about PH in COPD has been generated at sea level, with limited information associated with high altitude (HA). OBJECTIVES To assess the prevalence and severity of PH in COPD patients living in a HA city (2,640 m). METHODS Cross-sectional study in COPD patients with forced expiratory volume in the first second / forced vital capacity ratio (FEV1/FVC) post-bronchodilator <0,7. Transthoracic echocardiography (TTE), spirometry, carbon monoxide diffusing capacity, and arterial blood gasses tests were performed. Patients were classified according to the severity of airflow limitation. PH was defined by TTE as an estimated systolic pulmonary artery pressure (sPAP) > 36 mmHg or indirect PH signs; severe PH as sPAP > 60 mmHg; and disproportionate PH as an sPAP > 60 mmHg with non-severe airflow limitation (FEV1 > 50% predicted). RESULTS We included 176 COPD patients. The overall estimated prevalence of PH was 56.3% and the likelihood of having PH increased according to airflow-limitation severity: mild (31.6%), moderate (54.9%), severe (59.6%) and very severe (77.8%) (p = 0.038). The PH was severe in 7.3% and disproportionate in 3.4% of patients. CONCLUSIONS The estimated prevalence of PH in patients with COPD at HA is high, particularly in patients with mild to moderate airflow limitation, and greater than that described for COPD patients at low altitude. These results suggest a higher risk of developing PH for COPD patients living at HA compared to COPD patients with similar airflow limitation living at low altitude.
Collapse
Affiliation(s)
- C Aguirre-Franco
- Fundación Neumológica Colombiana. Bogotá, Colombia; Universidad de La Sabana. Chía, Colombia.
| | - C A Torres-Duque
- Fundación Neumológica Colombiana. Bogotá, Colombia; Universidad de La Sabana. Chía, Colombia
| | - G Salazar
- Fundación Cardioinfantil - Instituto de Cardiología. Bogotá, Colombia
| | - A Casas
- Fundación Neumológica Colombiana. Bogotá, Colombia; Universidad de La Sabana. Chía, Colombia
| | - C Jaramillo
- Universidad de La Sabana. Chía, Colombia; Fundación Clínica Shaio. Bogotá, Colombia
| | - M Gonzalez-Garcia
- Fundación Neumológica Colombiana. Bogotá, Colombia; Universidad de La Sabana. Chía, Colombia
| |
Collapse
|
17
|
Yang S, Zhang T, Ge Y, Yin L, Pu Y, Liang G. Inhalation exposure to polystyrene nanoplastics induces chronic obstructive pulmonary disease-like lung injury in mice through multi-dimensional assessment. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 347:123633. [PMID: 38423272 DOI: 10.1016/j.envpol.2024.123633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/10/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
Nanoplastics are widely distributed in indoor and outdoor air and can be easily inhaled into human lungs. However, limited studies have investigated the impact of nanoplastics on inhalation toxicities, especially on the initiation and progression of chronic obstructive pulmonary disease (COPD). To fill the gap, the present study used oronasal aspiration to develop mice models. Mice were exposed to polystyrene nanoplastics (PS-NPs) at three concentrations, as well as the corresponding controls, for acute, subacute, and subchronic exposure. As a result, PS-NPs could accumulate in exposed mice lungs and influence lung organ coefficient. Besides, PS-NPs induced local and systemic oxidative stress, inflammation, and protease-antiprotease imbalance, resulting in decreased respiratory function and COPD-like lesions. Meanwhile, PS-NPs could trigger the subcellular mechanism to promote COPD development by causing mitochondrial dysfunctions and endoplasmic reticulum (ER) stress. Mechanistically, ferroptosis played an important role in the COPD-like lung injury induced by PS-NPs. In summary, the present study comprehensively and systematically indicates that PS-NPs can damage human respiratory health and increase the risk for COPD.
Collapse
Affiliation(s)
- Sheng Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China; Institute of Biomedical Devices (Suzhou), Southeast University, Suzhou, 215163, China.
| | - Tianyi Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China; Institute of Biomedical Devices (Suzhou), Southeast University, Suzhou, 215163, China.
| | - Yiling Ge
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China; Institute of Biomedical Devices (Suzhou), Southeast University, Suzhou, 215163, China.
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China.
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China.
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|
18
|
Wang K, Gao X, Yang H, Tian H, Zhang Z, Wang Z. Transcriptome analysis on pulmonary inflammation between periodontitis and COPD. Heliyon 2024; 10:e28828. [PMID: 38601631 PMCID: PMC11004760 DOI: 10.1016/j.heliyon.2024.e28828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 04/12/2024] Open
Abstract
Objective The aim of this study is to investigate the correlation between periodontal disease and chronic obstructive pulmonary disease (COPD) from the perspective of gene regulation, as well as the inflammatory pathways involved. Methods Forty C57BL/6 mice were randomly divided into four groups: control group, chronic periodontitis (CP) group, COPD group, and CP&COPD group. Lung tissue samples were selected for messenger ribonucleic acid (mRNA) sequencing analysis, and differential genes were screened out. Gene enrichment analysis was carried out, and then crosstalk gene enrichment analysis was conducted to explore the pathogenesis related to periodontal disease and COPD. Results Results of enrichment analysis showed that the differentially expressed genes (DEGs) in the CP group were concentrated in response to bacterial origin molecules. The DEGs in the COPD group gene were enriched in positive regulation of B cell activation. The DEGs in the CP&COPD group were concentrated in neutrophil extravasation and neutrophil migration. The mice in the three experimental groups had 19 crosstalk genes, five of which were key genes. Conclusions Lcn2, S100a8, S100a9, Irg1, Clec4d are potential crossover genes of periodontal disease and COPD. Lcn2, S100a8, S100a9 are correlated with neutrophils in both diseases. Irg1 and Clec4d may bind to receptors on the surface of lymphocytes to produce cytokines and activate inflammatory pathways, this requires further research.
Collapse
Affiliation(s)
- Kaili Wang
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Department of Stomatology, Beijing You 'an Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Gao
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Hongjia Yang
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Huan Tian
- Changsha Stomatological Hospital, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Zheng Zhang
- Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, China
| | - Zuomin Wang
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Luo L, Tang J, Du X, Li N. Chronic obstructive pulmonary disease and the airway microbiome: A review for clinicians. Respir Med 2024; 225:107586. [PMID: 38460708 DOI: 10.1016/j.rmed.2024.107586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 12/30/2023] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a complex heterogeneous disease characterized by progressive airflow limitation and chronic inflammation. The progressive development and long-term repeated acute exacerbation of COPD make many patients still unable to control the deterioration of the disease after active treatment, and even eventually lead to death. An increasing number of studies have shown that the occurrence and development of COPD are closely related to the composition and changes of airway microbiome. This article reviews the interaction between COPD and airway microbiome, the potential mechanisms of interaction, and the treatment methods related to microbiome. We elaborated the internal correlation between airway microbiome and different stages of COPD, inflammatory endotypes, glucocorticoid and antibiotic treatment, analyze the pathophysiological mechanisms such as the "vicious cycle" hypothesis, abnormal inflammation-immune response of the host and the "natural selection" of COPD to airway microbiome, introduce the treatment of COPD related to microbiome and emphasize the predictive value of airway microbiome for the progression, exacerbation and prognosis of COPD, as well as the guiding role for clinical management of patients, in order to provide a new perspective for exploring the pathogenesis of COPD, and also provide clues and guidance for finding new treatment targets.
Collapse
Affiliation(s)
- Lingxin Luo
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Junli Tang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Xianzhi Du
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Na Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China.
| |
Collapse
|
20
|
Marriott E, Singanayagam A, El-Awaisi J. Inflammation as the nexus: exploring the link between acute myocardial infarction and chronic obstructive pulmonary disease. Front Cardiovasc Med 2024; 11:1362564. [PMID: 38450367 PMCID: PMC10915015 DOI: 10.3389/fcvm.2024.1362564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 02/01/2024] [Indexed: 03/08/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD), particularly following acute exacerbations (AE-COPD), significantly heightens the risks and mortality associated with acute myocardial infarction (AMI). The intersection of COPD and AMI is characterised by a considerable overlap in inflammatory mechanisms, which play a crucial role in the development of both conditions. Although extensive research has been conducted on individual inflammatory pathways in AMI and COPD, the understanding of thrombo-inflammatory crosstalk in comorbid settings remains limited. The effectiveness of various inflammatory components in reducing AMI infarct size or slowing COPD progression has shown promise, yet their efficacy in the context of comorbidity with COPD and AMI is not established. This review focuses on the critical importance of both local and systemic inflammation, highlighting it as a key pathophysiological connection between AMI and COPD/AE-COPD.
Collapse
Affiliation(s)
- Eloise Marriott
- Microcirculation Research Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Aran Singanayagam
- MRC Centre for Molecular Bacteriology & Infection, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Juma El-Awaisi
- Microcirculation Research Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
21
|
Zhang Y, Tan X, Hu S, Cui Z, Chen W. Relationship Between Systemic Immune-Inflammation Index and Risk of Respiratory Failure and Death in COPD: A Retrospective Cohort Study Based on the MIMIC-IV Database. Int J Chron Obstruct Pulmon Dis 2024; 19:459-473. [PMID: 38404653 PMCID: PMC10888109 DOI: 10.2147/copd.s446364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/26/2024] [Indexed: 02/27/2024] Open
Abstract
Purpose Chronic obstructive pulmonary disease (COPD) concurrent with respiratory failure (RF) is devastating, and may result in death and disability. Systemic immune-inflammation index (SII) is a new prognostic biomarker linked to unfavorable outcomes of acute coronary syndrome, ischemic stroke, and heart failure. Nonetheless, its role in COPD is rarely investigated. Consequently, this study intends to assess the accuracy of SII in predicting the prognosis of COPD. Patients and Methods The clinical information was retrospectively acquired from the Medical Information Mart for Intensive Care-IV database. The outcomes encompassed the incidence of RF and mortality. The relationship between different SII and outcomes was examined utilizing the Cox proportional-hazards model and restricted cubic splines. Kaplan-Meier analysis was employed for all-cause mortality. Results The present study incorporated 1653 patients. During hospitalization, 697 patients (42.2%) developed RF, and 169 patients (10.2%) died. And 637 patients (38.5%) died during long-term follow-up. Higher SII increased the risk of RF (RF: HR: 1.19, 95% CI 1.12-1.28, P<0.001), in-hospital mortality (HR: 1.22, 95% CI 1.07-1.39, P=0.003), and long-term follow-up mortality (HR: 1.12, 95% CI 1.05-1.19, P<0.001). Kaplan-Meier analysis suggested a significantly elevated risk of all-cause death (log-rank P<0.001) in patients with higher SII, especially during the short-term follow-up period of 21 days. Conclusion SII is closely linked to an elevated risk of RF and death in COPD patients. It appears to be a potential predictor of the prognosis of COPD patients, which is helpful for the risk stratification of this population. However, more prospective studies are warranted to consolidate our conclusion.
Collapse
Affiliation(s)
- Ye Zhang
- Department of General Medicine, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, People’s Republic of China
| | - Xiaoli Tan
- Department of Respiratory Medicine, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, People’s Republic of China
| | - Shiyu Hu
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Jiaxing, Zhejiang, People’s Republic of China
| | - Zhifang Cui
- Department of Respiratory Medicine, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Wenyu Chen
- Department of Respiratory Medicine, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, People’s Republic of China
| |
Collapse
|
22
|
Li Z, Wang H, Yin Y. Peripheral inflammation is a potential etiological factor in Alzheimer's disease. Rev Neurosci 2024; 35:99-120. [PMID: 37602685 DOI: 10.1515/revneuro-2023-0049] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/27/2023] [Indexed: 08/22/2023]
Abstract
Peripheral inflammation could constitute a risk factor for AD. This review summarizes the research related to peripheral inflammation that appears to have a relationship with Alzheimer's disease. We find there are significant associations between AD and peripheral infection induced by various pathogens, including herpes simplex virus type 1, cytomegalovirus, Epstein-Barr virus, human immunodeficiency virus, severe acute respiratory syndrome coronavirus 2, Porphyromonas gingivalis, Helicobacter pylori, and Toxoplasma gondii. Chronic inflammatory diseases are also reported to contribute to the pathophysiology of AD. The mechanisms by which peripheral inflammation affects the pathophysiology of AD are complex. Pathogen-derived neurotoxic molecule composition, disrupted BBB, and dysfunctional neurogenesis may all play a role in peripheral inflammation, promoting the development of AD. Anti-pathogenic medications and anti-inflammatory treatments are reported to decrease the risk of AD. Studies that could improve understanding the associations between AD and peripheral inflammation are needed. If our assumption is correct, early intervention against inflammation may be a potential method of preventing and treating AD.
Collapse
Affiliation(s)
- Ziyuan Li
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Yangpu District, Shanghai 200092, China
| | - Hui Wang
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Yangpu District, Shanghai 200092, China
| | - Yafu Yin
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Yangpu District, Shanghai 200092, China
| |
Collapse
|
23
|
Patowary P, Pathak MP, Barbhuiya PA, Karmakar S, Chattopadhyay P, Zaman K. Oleoresin Capsicum (OC) Spray: An Assessment of Respiratory Health and its Management Following Accidental and Deliberate Exposures. Curr Top Med Chem 2024; 24:1542-1556. [PMID: 38778617 DOI: 10.2174/0115680266298811240514061433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/09/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024]
Abstract
Introduced into law enforcement in 1976, the oleoresin capsicum (OC) spray has been labeled as one of the most significant and radical developments in law enforcement. However, epidemiological research on OC health effects is deficient, receiving little public support. The major responses to acute exposure to OC spray can be found in the pulmonary system. The molecular mechanism(s) involved in the action of capsaicinoids, the active constituents in OC, are complex cascades of reactions which end up in necrosis or apoptosis. OC may also damage and deplete biological redox systems in the epithelial lining fluids and within cells and mitochondria, modifying structural proteins and nucleic acids and leading to enzyme inactivation. Since there are no characteristic laboratory tests available for identification or confirmation of OC exposure, and on the basis of prevailing data, reassessment of the health risks of OC exposures in vulnerable populations and in-depth study of the molecular mechanics of receptors is the need of the hour for the development of effective countermeasures. This review aims to consider evidence for adverse effects of OC spray used in ways comparable to their application by law enforcement personnel and civilians, with possible treatment recommendations that are precedent for improved management.
Collapse
Affiliation(s)
- Pompy Patowary
- Division of Pharmaceutical Technology, Defence Research Laboratory, Tezpur, 784 001, Assam, India
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786 004, Assam, India
| | - Manash Pratim Pathak
- Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India
- Centre for Research on Ethnomedicine, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India
| | - Pervej Alom Barbhuiya
- Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India
- Centre for Research on Ethnomedicine, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India
| | - Sanjeev Karmakar
- Division of Pharmaceutical Technology, Defence Research Laboratory, Tezpur, 784 001, Assam, India
| | - Pronobesh Chattopadhyay
- Division of Pharmaceutical Technology, Defence Research Laboratory, Tezpur, 784 001, Assam, India
| | - Kamaruz Zaman
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786 004, Assam, India
| |
Collapse
|
24
|
Lee Y, Song J, Jeong Y, Choi E, Ahn C, Jang W. Meta-analysis of single-cell RNA-sequencing data for depicting the transcriptomic landscape of chronic obstructive pulmonary disease. Comput Biol Med 2023; 167:107685. [PMID: 37976829 DOI: 10.1016/j.compbiomed.2023.107685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/17/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a respiratory disease characterized by airflow limitation and chronic inflammation of the lungs that is a leading cause of death worldwide. Since the complete pathological mechanisms at the single-cell level are not fully understood yet, an integrative approach to characterizing the single-cell-resolution landscape of COPD is required. To identify the cell types and mechanisms associated with the development of COPD, we conducted a meta-analysis using three single-cell RNA-sequencing datasets of COPD. Among the 154,011 cells from 16 COPD patients and 18 healthy subjects, 17 distinct cell types were observed. Of the 17 cell types, monocytes, mast cells, and alveolar type 2 cells (AT2 cells) were found to be etiologically implicated in COPD based on genetic and transcriptomic features. The most transcriptomically diversified states of the three etiological cell types showed significant enrichment in immune/inflammatory responses (monocytes and mast cells) and/or mitochondrial dysfunction (monocytes and AT2 cells). We then identified three chemical candidates that may potentially induce COPD by modulating gene expression patterns in the three etiological cell types. Overall, our study suggests the single-cell level mechanisms underlying the pathogenesis of COPD and may provide information on toxic compounds that could be potential risk factors for COPD.
Collapse
Affiliation(s)
- Yubin Lee
- Department of Life Sciences, Dongguk University, Seoul, 04620, Republic of Korea.
| | - Jaeseung Song
- Department of Life Sciences, Dongguk University, Seoul, 04620, Republic of Korea.
| | - Yeonbin Jeong
- Department of Life Sciences, Dongguk University, Seoul, 04620, Republic of Korea.
| | - Eunyoung Choi
- Department of Life Sciences, Dongguk University, Seoul, 04620, Republic of Korea.
| | - Chulwoo Ahn
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Wonhee Jang
- Department of Life Sciences, Dongguk University, Seoul, 04620, Republic of Korea.
| |
Collapse
|
25
|
Li J, Chen R, Yu X, Liu H. Guidelines of integrated Chinese and western medicine for diagnosis and treatment of chronic obstructive pulmonary disease (2022). J Evid Based Med 2023; 16:565-580. [PMID: 38130086 DOI: 10.1111/jebm.12578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023]
Abstract
Chronic obstructive pulmonary disease (COPD), with high prevalence rate, mortality, disability rate, and heavy disease burden, has become a critical chronic disease seriously threatening public health worldwide. Traditional Chinese medicine and Western medicine both have shown advantages in diagnosing and treating COPD, which has been widely applied in the clinics. In order to improve the diagnostic and treatment level for COPD with integrated traditional Chinese and Western medicine, the Guidelines of Integrated Chinese and Western Medicine for Diagnosis and Treatment of COPD were developed by the Internal Medicine Committee of the World Federation of Chinese Medicine Societies. First, a multidisciplinary working group was established. Development methods and processes of international clinical practice guidelines were adopted in the whole research. In final, a total of 13 recommendations for the diagnosis and treatment of COPD were established based on available evidence with the best quality. Meanwhile, characteristics of integrated traditional Chinese and Western medicine in treating COPD were taken into account with pros and cons of each intervention. The guidelines could be used as a reference for physicians in respiratory medicine departments (traditional Chinese medicine, integrated traditional Chinese and Western medicine, and Western medicine) at various levels of medical institutions in their diagnosis and treatment.
Collapse
Affiliation(s)
- Jiansheng Li
- Co-Construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Department of Respiratory Diseases, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Rongchang Chen
- Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, China
| | - Xueqing Yu
- Co-Construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of China, Henan University of Chinese Medicine, Zhengzhou, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Department of Respiratory Diseases, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Huiguo Liu
- Department of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
Qin Y, Zhai J, Yang J, Li H, Tian Y, Liu X, Zhao P, Li J. Effective-component compatibility of Bufei Yishen formula alleviates chronic obstructive pulmonary disease inflammation by regulating GSK3β-mediated NLRP3 inflammasome activation. Biomed Pharmacother 2023; 168:115614. [PMID: 37862971 DOI: 10.1016/j.biopha.2023.115614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/19/2023] [Accepted: 09/27/2023] [Indexed: 10/22/2023] Open
Abstract
Glycogen synthase kinase 3β (GSK3β) has been associated with sensing many different stimuli to trigger the NLRP3 inflammasome, which plays a crucial role in promoting the inflammatory response in diseases, including chronic obstructive pulmonary disease (COPD). Bufei Yishen formula (BYF), a traditional Chinese herbal medicine, has beneficial effects on COPD. Effective-component compatibility of BYF (ECC-BYF), optimized from BYF, is equally effective as BYF in inhibiting COPD inflammation. However, the exact mechanism by which ECC-BYF regulates the activation of NLRP3 inflammasome to inhibit COPD inflammation remains unclear. Hence, we investigated the mechanisms underlying the alleviation of COPD inflammation by ECC-BYF through the inhibition of GSK3β-mediated NLRP3 inflammasome activation by experimental rat model of COPD and lipopolysaccharide/adenosine triphosphate (LPS/ATP) induced macrophages. The data showed that ECC-BYF significantly improved the lung function, attenuated histopathological damage, and alleviated inflammatory cell infiltration and alveolar destruction. Further, it significantly inhibited inflammatory cytokine production and downregulated the phosphorylation of GSK3β by inhibiting the activation of NLRP3 inflammasome in the rat model of COPD. Moreover, ECC-BYF suppressed the activation of the NLRP3 inflammasome by increasing the phosphorylation at serine 9 and decreasing the phosphorylation at tyrosine 216 of GSK3β, followed by the inhibition of IL-1β secretion in macrophages. Together, ECC-BYF effectively ameliorates COPD by suppressing inflammation, which is dependent on the regulation of GSK3β-mediated NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Yanqin Qin
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Jiena Zhai
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Jingfan Yang
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Haibo Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Yange Tian
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Xuefang Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Peng Zhao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan and Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou 450046, Henan Province, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, Henan Province, China.
| |
Collapse
|
27
|
Shen Y, Chen L, Chen J, Qin J, Wang T, Wen F. Mitochondrial damage-associated molecular patterns in chronic obstructive pulmonary disease: Pathogenetic mechanism and therapeutic target. J Transl Int Med 2023; 11:330-340. [PMID: 38130648 PMCID: PMC10732348 DOI: 10.2478/jtim-2022-0019] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common inflammatory airway disease characterized by enhanced inflammation. Recent studies suggest that mitochondrial damage-associated molecular patterns (DAMPs) may play an important role in the regulation of inflammation and are involved in a serial of inflammatory diseases, and they may also be involved in COPD. This review highlights the potential role of mitochondrial DAMPs during COPD pathogenesis and discusses the therapeutic potential of targeting mitochondrial DAMPs and their related signaling pathways and receptors for COPD. Research progress on mitochondrial DAMPs may enhance our understanding of COPD inflammation and provide novel therapeutic targets.
Collapse
Affiliation(s)
- Yongchun Shen
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, Chengdu610041, Sichuan Province, China
| | - Lei Chen
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, Chengdu610041, Sichuan Province, China
| | - Jun Chen
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, Chengdu610041, Sichuan Province, China
| | - Jiangyue Qin
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, Chengdu610041, Sichuan Province, China
| | - Tao Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, Chengdu610041, Sichuan Province, China
| | - Fuqiang Wen
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, Chengdu610041, Sichuan Province, China
| |
Collapse
|
28
|
Hadzic S, Wu CY, Gredic M, Pak O, Loku E, Kraut S, Kojonazarov B, Wilhelm J, Brosien M, Bednorz M, Seimetz M, Günther A, Kosanovic D, Sommer N, Warburton D, Li X, Grimminger F, Ghofrani HA, Schermuly RT, Seeger W, El Agha E, Bellusci S, Weissmann N. Fibroblast growth factor 10 reverses cigarette smoke- and elastase-induced emphysema and pulmonary hypertension in mice. Eur Respir J 2023; 62:2201606. [PMID: 37884305 PMCID: PMC10632559 DOI: 10.1183/13993003.01606-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 08/28/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND COPD is an incurable disease and a leading cause of death worldwide. In mice, fibroblast growth factor (FGF)10 is essential for lung morphogenesis, and in humans, polymorphisms in the human FGF10 gene correlate with an increased susceptibility to develop COPD. METHODS We analysed FGF10 signalling in human lung sections and isolated cells from healthy donor, smoker and COPD lungs. The development of emphysema and PH was investigated in Fgf10+/- and Fgfr2b+/- (FGF receptor 2b) mice upon chronic exposure to cigarette smoke. In addition, we overexpressed FGF10 in mice following elastase- or cigarette smoke-induced emphysema and pulmonary hypertension (PH). RESULTS We found impaired FGF10 expression in human lung alveolar walls and in primary interstitial COPD lung fibroblasts. In contrast, FGF10 expression was increased in large pulmonary vessels in COPD lungs. Consequently, we identified impaired FGF10 signalling in alveolar walls as an integral part of the pathomechanism that leads to emphysema and PH development: mice with impaired FGF10 signalling (Fgf10+/- and Fgfr2b+/- ) spontaneously developed lung emphysema, PH and other typical pathomechanistic features that generally arise in response to cigarette smoke exposure. CONCLUSION In a therapeutic approach, FGF10 overexpression successfully restored lung alveolar and vascular structure in mice with established cigarette smoke- and elastase-induced emphysema and PH. FGF10 treatment triggered an initial increase in the number of alveolar type 2 cells that gradually returned to the basal level when the FGF10-mediated repair process progressed. Therefore, the application of recombinant FGF10 or stimulation of the downstream signalling cascade might represent a novel therapeutic strategy in the future.
Collapse
Affiliation(s)
- Stefan Hadzic
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Cheng-Yu Wu
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Marija Gredic
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Oleg Pak
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Edma Loku
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Simone Kraut
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Baktybek Kojonazarov
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
- Institute for Lung Health (ILH), Justus-Liebig-University, Giessen, Germany
| | - Jochen Wilhelm
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
- Institute for Lung Health (ILH), Justus-Liebig-University, Giessen, Germany
| | - Monika Brosien
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Mariola Bednorz
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Michael Seimetz
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Andreas Günther
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Djuro Kosanovic
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Natascha Sommer
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - David Warburton
- Children's Hospital Los Angeles, Los Angeles, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, P.R. China
| | - Friedrich Grimminger
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Hossein A Ghofrani
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Ralph T Schermuly
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
- Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Elie El Agha
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
- Institute for Lung Health (ILH), Justus-Liebig-University, Giessen, Germany
| | - Saverio Bellusci
- Oujiang Laboratory (Zheijiang Laboratory for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
- Laboratory of Extracellular Matrix Remodelling, Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
- S. Bellusci and N. Weissmann contributed equally to this article as lead authors and supervised the work
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Giessen, Germany
- S. Bellusci and N. Weissmann contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
29
|
Yu Y, Yang A, He X, Wu B, Wu Y, Li Y, Nie S, Xu B, Wang H, Yu G. Soluble epoxide hydrolase deficiency attenuates airway inflammation in COPD via IRE1α/JNK/AP-1 signaling pathway. J Inflamm (Lond) 2023; 20:36. [PMID: 37915073 PMCID: PMC10621191 DOI: 10.1186/s12950-023-00361-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND Soluble Epoxide Hydrolase (sEH) metabolizes anti-inflammatory epoxyeicosatrienoic acids and critically affects airway inflammation in chronic obstructive pulmonary disease (COPD). Considering the excessive endoplasmic reticulum stress is associated with the earlier onset of COPD. The role of sEH and endoplasmic reticulum stress in the pathogenesis of COPD remains unknown. METHOD 16 weeks of cigarette-exposed mice were used to detect the relationship between sEH and endoplasmic reticulum stress in COPD. Human epithelial cells were used in vitro to determine the regulation mechanism of sEH in endoplasmic reticulum stress induced by cigarette smoke. RESULTS sEH deficiency helps reduce emphysema formation after smoke exposure by alleviating endoplasmic reticulum stress response. sEH deficiency effectively reverses the upregulation of phosphorylation IRE1α and JNK and the nuclear expression of AP-1, alleviating the secretion of inflammatory factors induced by cigarette smoke extract. Furthermore, the treatment with endoplasmic reticulum stress and IRE1α inhibitor downregulated cigarette smoke extract-induced sEH expression and the secretion of inflammatory factors. CONCLUSION sEH probably alleviates airway inflammatory response and endoplasmic reticulum stress via the IRE1α/JNK/AP-1 pathway, which might attenuate lung injury caused by long-term smoking and provide a new pharmacological target for preventing and treating COPD.
Collapse
Affiliation(s)
- Yue Yu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China
| | - Ailin Yang
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China
| | - Xin He
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China
| | - Bo Wu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China
| | - Yanjun Wu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China
| | - Yunxiao Li
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China
| | - Shan Nie
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China
| | - Bo Xu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China.
| | - Haoyan Wang
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China.
| | - Ganggang Yu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, No, 95 Yong An Road, Xichen District, Beijing, 100050, China.
| |
Collapse
|
30
|
Shen S, Xiao Y. Association Between C-Reactive Protein and Albumin Ratios and Risk of Mortality in Patients with Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2023; 18:2289-2303. [PMID: 37873518 PMCID: PMC10590598 DOI: 10.2147/copd.s413912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 09/14/2023] [Indexed: 10/25/2023] Open
Abstract
Purpose This study was to evaluate the association between C-reactive protein (CRP) to albumin (ALB) ratio (CAR) and the risk of mortality in patients with chronic obstructive pulmonary disease (COPD). Methods In this retrospective cohort study, clinical data were extracted from the National Institutes of Health National Health and Nutrition Examination Survey (NHANES) database between 2007 and 2010. Cox proportional hazard regressions were performed to assess the association between CAR and 5-year mortality in COPD patients. Subgroup analyses were applied to identify the consistency of the association based on the severity of COPD, gender, body mass index (BMI), smoking status, cardiovascular disease (CVD), and chronic kidney disease (CKD), and diabetes. The area under the curve (AUC) of the receiver operator characteristic (ROC) curve analysis was used to evaluate the predictive performance of CAR. Results A total of 1210 COPD patients were included, of which 110 COPD patients (9.09%) had 5-year mortality. The mean follow-up was 57.76 (0.33) months. A higher CAR was associated with an increased risk of 5-year mortality in COPD patients [hazard ratio (HR): 1.94, 95% confidence interval (CI): 1.07 to 3.50, P =0.029)]. Subgroup analysis showed that the association between CAR and mortality was especially suitable for COPD patients with mild COPD, in COPD patients who were still smoking, in COPD patients with BMI <=29.9 kg/m2, in COPD patients who were without CVD, in COPD patients who were without diabetes, and COPD patients who were without CKD. The AUCs of CAR for predicting 1-year, 3-year, and 5-year mortality in COPD patients were 0.735, 0.615, and 0.608, respectively. Conclusion CAR is strongly correlated with mortality in patients with COPD and CAR could be served as a prognostic biomarker for patients with COPD. This study may provide a promising prognostic biomarker for risk stratification and clinical management of patients with COPD.
Collapse
Affiliation(s)
- Shenghua Shen
- Department of Pulmonary and Critical Care Medicine, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, 213004, People’s Republic of China
| | - Yuan Xiao
- Department of Pulmonary and Critical Care Medicine, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, 213004, People’s Republic of China
| |
Collapse
|
31
|
Beykumül A, Ersoy Y, Gülbaş G, Neselioglu S. Can Blood Biomarkers Be Used to Assess Oxidative Stress in COPD Patients After Pulmonary Rehabilitation. Int J Chron Obstruct Pulmon Dis 2023; 18:2179-2186. [PMID: 37818173 PMCID: PMC10561608 DOI: 10.2147/copd.s400415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 08/21/2023] [Indexed: 10/12/2023] Open
Abstract
Purpose To determine the level of oxidative stress in the body due to pulmonary rehabilitation (PR) with thiols and disulfide and to investigate their relationship with indirect markers such as creatine kinase (CK), creatine kinase - MB (CK-MB), and lactate dehydrogenase (LDH), which show cell destruction. Patients and Methods Patients with chronic obstructive pulmonary disease (COPD) are included in inpatient and outpatient care. They were evaluated before and after for PR, and an exercise program was prescribed. In addition, native thiol (NT), total thiol (TT), disulfide (DS), LDH, CK, and CK-MB values were tested. Results The mean age of 21 patients was 63±7.31 years. Eleven of them were outpatients and 10 of them were inpatients. Most of the patients were male (M/F=20/1, 95.2/4.8%). There was a significant difference in pulmonary function tests (PFT), St. George Respiratory Questionnaire (SGRQ), and 1 repetition maximum (1RM) before and after the treatment (p<0.001). There was a correlation between PFT and 1RM upper extremity. While there was no significant difference between thiols and disulfide, according to GOLD scores, there was a significant difference in patients with level 3-MMRC. No correlation was found between LDH, CK, CK-MB, and thiols, DS. ΔCK was found to be associated with ΔDS, and ΔCK-MB with ΔNT, and ΔTT. Conclusion PR contributes to the antioxidant process by improving respiration and reducing oxidative stress. The decrease in LDH, CK with PR, increase in CK-MB, and correlation of CK with thiols and DS gave a different interpretation. In this case, it should be considered that oxidative stress may also be increased in people with high CK values.
Collapse
Affiliation(s)
- Ayşegül Beykumül
- Pulmonary Rehabilitation Unit, Turgut Ozal Medical Center, Inonu University, Malatya, Turkey
| | - Yuksel Ersoy
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Gazi Gülbaş
- Department of Chest Diseases, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Salim Neselioglu
- Department of Medical Biochemistry, Faculty of Medicine, Yildirim Beyazit University, Ankara, Turkey
| |
Collapse
|
32
|
Huang X, Li J, Cao W, Lyu J, Guo Y, Pei P, Xia Q, Du H, Chen Y, Ling Y, Kerosi R, Stevens R, Yang X, Chen J, Yu C, Chen Z, Li L. Association between fresh fruit consumption and the risk of chronic obstructive pulmonary disease-related hospitalization and death in Chinese adults: A prospective cohort study. Chin Med J (Engl) 2023; 136:2316-2323. [PMID: 37537725 PMCID: PMC10538915 DOI: 10.1097/cm9.0000000000002591] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Existing evidence suggests that fruit consumption is a significant influencing factor for chronic obstructive pulmonary disease (COPD), but this is unclear in the Chinese population. We examined the association of fresh fruit consumption with the risk of COPD-related hospitalization and death in a nationwide, population-based prospective cohort from China. METHODS Between 2004 and 2008, the China Kadoorie Biobank recruited >0.5 million adults aged 30 to 79 years from ten diverse regions across China. After excluding individuals diagnosed with major chronic diseases and prevalent COPD, the prospective analysis included 421,428 participants. Cox regression was used to calculate the hazard ratios (HRs) for the association between fresh fruit consumption and risk of COPD-related hospitalization and death, with adjustment for established and potential confounders. RESULTS During a mean follow-up of 10.9 years, 11,292 COPD hospitalization events and deaths were documented, with an overall incidence rate of 2.47/1000 person-years. Participants who consumed fresh fruit daily had a 22% lower risk of COPD-related hospitalization and death compared with non-consumers (HR = 0.78, 95% confidence interval [CI]: 0.71-0.87). The inverse association between fresh fruit consumption and COPD-related hospitalization and death was stronger among non-current smokers and participants with normal body mass index (BMI) (18.5 kg/m 2 ≤ BMI < 24.0 kg/m 2 ); the corresponding HRs for daily fresh fruit consumption were 0.78 (95% CI: 0.68-0.89) and 0.69 (95% CI: 0.59-0.79) compared with their counterparts, respectively. CONCLUSIONS High-frequency fruit consumption was associated with a lower risk of COPD in Chinese adults. Increasing fruit consumption, together with cigarette cessation and weight control, should be considered in the prevention and management of COPD.
Collapse
Affiliation(s)
- Xin Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China
| | - Jiachen Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China
| | - Weihua Cao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China
| | - Jun Lyu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing 100191, China
| | - Yu Guo
- National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences, National Center for Cardiovascular Diseases, Beijing 100037, China
| | - Pei Pei
- National Coordinate Center, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Qingmei Xia
- National Coordinate Center, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Huaidong Du
- Nuffield Department of Population Health, Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), University of Oxford, Oxford OX3 7LF, UK
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford OX3 7LF, UK
| | - Yiping Chen
- Nuffield Department of Population Health, Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), University of Oxford, Oxford OX3 7LF, UK
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford OX3 7LF, UK
| | - Yang Ling
- Nuffield Department of Population Health, Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), University of Oxford, Oxford OX3 7LF, UK
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford OX3 7LF, UK
| | - Rene Kerosi
- Nuffield Department of Population Health, Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), University of Oxford, Oxford OX3 7LF, UK
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford OX3 7LF, UK
| | - Rebecca Stevens
- Nuffield Department of Population Health, Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), University of Oxford, Oxford OX3 7LF, UK
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford OX3 7LF, UK
| | - Xujun Yang
- Maiji Center for Disease Control and Prevention, Tianshui, Gansu 741020, China
| | - Junshi Chen
- China National Center for Food Safety Risk Assessment, Beijing 100022, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing 100191, China
| | - Zhengming Chen
- Nuffield Department of Population Health, Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), University of Oxford, Oxford OX3 7LF, UK
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing 100191, China
| |
Collapse
|
33
|
Lu Y, Deng M, Yin Y, Hou G, Zhou X. Global Trends in Research Regarding Macrophages Associated with Chronic Obstructive Pulmonary Disease: A Bibliometric Analysis from 2011 to 2022. Int J Chron Obstruct Pulmon Dis 2023; 18:2163-2177. [PMID: 37810373 PMCID: PMC10558051 DOI: 10.2147/copd.s419634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/21/2023] [Indexed: 10/10/2023] Open
Abstract
Purpose Chronic obstructive pulmonary disease (COPD) is a prevalent respiratory condition characterized by chronic airway inflammation, where macrophages from the innate immune system may exert a pivotal influence. Our study aimed to summarize the present state of knowledge and to identify the focal points and emerging developments regarding macrophages associated with COPD through bibliometrics. Methods Publications regarding research on macrophages associated with COPD from January 1, 2011, to January 1, 2022, were retrieved from the Science Citation Index-Expanded (SCI-E) which is part of the Web of Science database. In total, 1521 publications were analyzed using bibliometric methodology. VOSviewer was used to analyze the annual publications, countries, institutions, authors, journals, and research hotspots. Results Based on the bibliometric analysis, publications relating to macrophages associated with COPD tended to increase from 2011 to 2022. The United States was the largest producer and most influential country in this field. Research during the past decade has focused on inflammation in the lungs. Most previous studies have mainly focused on the mechanisms that promote the initiation and progression of COPD. Macrophage-related oxidative stress and immunity, communication between macrophages and epithelial cells, and interventions for acute exacerbations have become the focus of more recent studies and will become a hot topic in the future. Conclusion Global research on macrophage-associated COPD has been growing rapidly in the past decade. The hot topics in this field gradually tended to shift focus from "inflammation" to "oxidative stress", "epithelial-cells", and "exacerbations". The significance of macrophages in coordinating immune responses, interacting with other cells, and exhibiting dysregulated capacities has attracted increasing attention to COPD pathogenesis. The adoption of new technologies may provide a more promising and comprehensive understanding of the specific role of macrophages in COPD in the future.
Collapse
Affiliation(s)
- Ye Lu
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Mingming Deng
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine, Beijing, People’s Republic of China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- National Clinical Research Center for Respiratory Diseases, Beijing, People’s Republic of China
| | - Yan Yin
- Department of Respiratory and Critical Care Medicine, First Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Gang Hou
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- National Center for Respiratory Medicine, Beijing, People’s Republic of China
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
- National Clinical Research Center for Respiratory Diseases, Beijing, People’s Republic of China
| | - Xiaoming Zhou
- Department of Respirology, Fuwai Hospital, National Center for Cardiovascular Diseases, Beijing, People’s Republic of China
| |
Collapse
|
34
|
Harrington JS, Ryter SW, Plataki M, Price DR, Choi AMK. Mitochondria in health, disease, and aging. Physiol Rev 2023; 103:2349-2422. [PMID: 37021870 PMCID: PMC10393386 DOI: 10.1152/physrev.00058.2021] [Citation(s) in RCA: 129] [Impact Index Per Article: 129.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023] Open
Abstract
Mitochondria are well known as organelles responsible for the maintenance of cellular bioenergetics through the production of ATP. Although oxidative phosphorylation may be their most important function, mitochondria are also integral for the synthesis of metabolic precursors, calcium regulation, the production of reactive oxygen species, immune signaling, and apoptosis. Considering the breadth of their responsibilities, mitochondria are fundamental for cellular metabolism and homeostasis. Appreciating this significance, translational medicine has begun to investigate how mitochondrial dysfunction can represent a harbinger of disease. In this review, we provide a detailed overview of mitochondrial metabolism, cellular bioenergetics, mitochondrial dynamics, autophagy, mitochondrial damage-associated molecular patterns, mitochondria-mediated cell death pathways, and how mitochondrial dysfunction at any of these levels is associated with disease pathogenesis. Mitochondria-dependent pathways may thereby represent an attractive therapeutic target for ameliorating human disease.
Collapse
Affiliation(s)
- John S Harrington
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York, United States
| | | | - Maria Plataki
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York, United States
| | - David R Price
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York, United States
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, New York-Presbyterian Hospital/Weill Cornell Medical Center, Weill Cornell Medicine, New York, New York, United States
| |
Collapse
|
35
|
Lin S, Liu C, Sun J, Guan Y. RNA-Sequencing and Bioinformatics Analysis of Exosomal Long Noncoding RNAs Revealed a Novel ceRNA Network in Stable COPD. Int J Chron Obstruct Pulmon Dis 2023; 18:1995-2007. [PMID: 37720876 PMCID: PMC10503524 DOI: 10.2147/copd.s414901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/24/2023] [Indexed: 09/19/2023] Open
Abstract
Purpose Exosomes are able to exchange their bioactive RNA cargo to recipient cells. In COPD, exosomes can be controlled and engineered for its use as targeted diagnostic and therapeutic tool. Our study explored novel lncRNAs and mRNAs in plasma exosomes that could be involved in the pathogenesis of COPD. Methods High-throughput sequencing was conducted to detect the alterations in the expression of exosomal lncRNAs and mRNAs. Gene ontology (GO) functional analyses and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were used to determine the significant functions and pathways associated with differentially expressed (DE) lncRNAs. The mRNA expression profile dataset, GSE76925, and microRNA expression profile dataset, GSE70080, were obtained from the GEO database. Venn diagrams were used to find common DE mRNAs between my mRNAs dataset and GSE76925. These common DEGs were subjected to PPI analyses to identify Hub genes. Subsequently, Venn diagrams were used to identify common genes between the target genes of DE-miRNAs and Hub genes as well as DE-miRNAs and my lncRNAs dataset. Finally, a lncRNA-miRNA-mRNA co-expression network was constructed by prediction using proprietary software. The lncRNA and mRNA expressions were then validated by quantitative reverse-transcription polymerase chain reaction (qRT-PCR). Results We identified 1578 differentially regulated lncRNAs and 3071 differentially regulated mRNAs. GO and KEGG pathway analyses suggested that the DE lncRNAs are involved in the pathogenesis of COPD. A lncRNA-miRNA-mRNA meshwork was established to predict the potential interactions among these RNAs. RP3-329A5.8 and MRPS11 expression was then subjected to qRT-PCR for validation. Correlations between MRPS11 and clinic-pathological features were explored. Conclusion Our study provided a set of lncRNAs and mRNAs that may be involved in the pathogenesis of COPD, thereby highlighting the need for further research on both diagnostic biomarkers and molecular mechanisms.
Collapse
Affiliation(s)
- Shan Lin
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Caihong Liu
- Department of Clinical Laboratory, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Jingting Sun
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Yinghui Guan
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
36
|
Ren J, Chen W, Zhong Z, Wang N, Chen X, Yang H, Li J, Tang P, Fan Y, Lin F, Bai C, Wu J. Bronchoalveolar Lavage Fluid from Chronic Obstructive Pulmonary Disease Patients Increases Neutrophil Chemotaxis Measured by a Microfluidic Platform. MICROMACHINES 2023; 14:1740. [PMID: 37763903 PMCID: PMC10537285 DOI: 10.3390/mi14091740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/25/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a persistent and progressive respiratory disorder characterized by expiratory airflow limitation caused by chronic inflammation. Evidence has shown that COPD is correlated with neutrophil chemotaxis towards the airways, resulting in neutrophilic airway inflammation. This study aimed to evaluate neutrophil chemotaxis in bronchoalveolar lavage fluid (BALF) from COPD patients using a high-throughput nine-unit microfluidic platform and explore the possible correlations between neutrophil migratory dynamics and COPD development. The results showed that BALF from COPD patients induced stronger neutrophil chemotaxis than the Control BALF. Our results also showed that the chemotactic migration of neutrophils isolated from the blood of COPD patients was not significantly different from neutrophils from healthy controls, and neutrophil migration in three known chemoattractants (fMLP, IL-8, and LTB4) was not affected by glucocorticoid treatment. Moreover, comparison with clinical data showed a trend of a negative relationship between neutrophil migration chemotactic index (C. I.) in COPD BALF and patient's spirometry data, suggesting a potential correlation between neutrophil migration and the severity of COPD. The present study demonstrated the feasibility of using the microfluidic platform to assess neutrophil chemotaxis in COPD pathogenesis, and it may serve as a potential marker for COPD evaluation in the future.
Collapse
Affiliation(s)
- Jiaqi Ren
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenfang Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen University General Hospital, Shenzhen 518055, China
| | - Zhicheng Zhong
- Department of Pulmonary and Critical Care Medicine, Shenzhen University General Hospital, Shenzhen 518055, China
| | - Ning Wang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Xi Chen
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Hui Yang
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jing Li
- Department of Pulmonary and Critical Care Medicine, Shenzhen University General Hospital, Shenzhen 518055, China
| | - Ping Tang
- Department of Pulmonary and Critical Care Medicine, Shenzhen University General Hospital, Shenzhen 518055, China
| | - Yanping Fan
- School of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Francis Lin
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Changqing Bai
- Department of Pulmonary and Critical Care Medicine, Shenzhen University General Hospital, Shenzhen 518055, China
| | - Jiandong Wu
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
37
|
Fan LC, McConn K, Plataki M, Kenny S, Williams NC, Kim K, Quirke JA, Chen Y, Sauler M, Möbius ME, Chung KP, Area Gomez E, Choi AM, Xu JF, Cloonan SM. Alveolar type II epithelial cell FASN maintains lipid homeostasis in experimental COPD. JCI Insight 2023; 8:e163403. [PMID: 37606038 PMCID: PMC10543729 DOI: 10.1172/jci.insight.163403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 07/10/2023] [Indexed: 08/23/2023] Open
Abstract
Alveolar epithelial type II (AEC2) cells strictly regulate lipid metabolism to maintain surfactant synthesis. Loss of AEC2 cell function and surfactant production are implicated in the pathogenesis of the smoking-related lung disease chronic obstructive pulmonary disease (COPD). Whether smoking alters lipid synthesis in AEC2 cells and whether altering lipid metabolism in AEC2 cells contributes to COPD development are unclear. In this study, high-throughput lipidomic analysis revealed increased lipid biosynthesis in AEC2 cells isolated from mice chronically exposed to cigarette smoke (CS). Mice with a targeted deletion of the de novo lipogenesis enzyme, fatty acid synthase (FASN), in AEC2 cells (FasniΔAEC2) exposed to CS exhibited higher bronchoalveolar lavage fluid (BALF) neutrophils, higher BALF protein, and more severe airspace enlargement. FasniΔAEC2 mice exposed to CS had lower levels of key surfactant phospholipids but higher levels of BALF ether phospholipids, sphingomyelins, and polyunsaturated fatty acid-containing phospholipids, as well as increased BALF surface tension. FasniΔAEC2 mice exposed to CS also had higher levels of protective ferroptosis markers in the lung. These data suggest that AEC2 cell FASN modulates the response of the lung to smoke by regulating the composition of the surfactant phospholipidome.
Collapse
Affiliation(s)
- Li-Chao Fan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Keith McConn
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Maria Plataki
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA
| | - Sarah Kenny
- School of Medicine, Trinity Biomedical Sciences Institute, and
| | | | - Kihwan Kim
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | | | - Yan Chen
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Maor Sauler
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Kuei-Pin Chung
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- Department of Laboratory Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Estela Area Gomez
- Division of Neuromuscular Medicine, Department of Neurology, Columbia University Irving Medical Center, Neurological Institute, New York, New York, USA
- Center for Biological Research “Margarita Salas”, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Augustine M.K. Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA
| | - Jin-Fu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Suzanne M. Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- School of Medicine, Trinity Biomedical Sciences Institute, and
| |
Collapse
|
38
|
Ye C, Yuan L, Wu K, Shen B, Zhu C. Association between systemic immune-inflammation index and chronic obstructive pulmonary disease: a population-based study. BMC Pulm Med 2023; 23:295. [PMID: 37563621 PMCID: PMC10416535 DOI: 10.1186/s12890-023-02583-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND The Systemic Immune-Inflammation Index (SII) is a quantitative measurement of the systemic immune-inflammatory response in the human body. The SII has been shown to have prognostic value in various clinical settings, including critical illness, sepsis, and cancer. Its role in chronic obstructive pulmonary disease (COPD) remains unclear and requires further investigation. METHODS We analyzed demographic data from 16,636 participants in the National Health and Nutrition Examination Survey. Logistic regression analysis was performed to assess the correlation between COPD, lung function, chronic respiratory symptoms and SII. We used Cox proportional hazards (PH) model to analyze the relationship between SII and mortality in COPD patients and healthy individuals. We used propensity score matching (PSM) method to match the COPD population with similar baseline levels with the normal population to further analyze the correlation between SII and COPD. RESULTS We recruited 16,636 participants, ages 40 and above, for the study. A multivariable logistic regression analysis revealed that a higher SII level was independently associated with an elevated likelihood of COPD (Odds Ratio (OR) = 1.449; 95% Confidence Interval (CI): 1.252-1.676, P < 0.0001) after controlling for all other factors. Results of subgroup analysis showed a significant positive correlation between SII and COPD in different age groups, gender, Body Mass Index, smoking status, and those with a history of hypertension. The SII index had positive correlation with COPD after PSM (OR = 1.673; 95%CI: 1.443-1.938). After full adjustment, an increase in the SII is associated with a higher all-cause mortality rate. The hazard ratio (HR) with a 95% CI in the general population, COPD patients, and healthy individuals are 1.161 (1.088, 1.239), 1.282 (1.060, 1.550), and 1.129 (1.055, 1.207), respectively. CONCLUSIONS Higher SII levels are linked to higher prevalence of COPD. COPD patients with a higher SII levels have a higher risk of all-cause mortality. Additional large-scale, long-term studies are necessary to confirm these results.
Collapse
Affiliation(s)
- Chenglin Ye
- Department of Clinical Laboratory, institute of translational medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Li Yuan
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, PR China
| | - Bingzheng Shen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Chengliang Zhu
- Department of Clinical Laboratory, institute of translational medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China.
| |
Collapse
|
39
|
Ma R, Su H, Jiao K, Liu J. Association Between IL-17 and Chronic Obstructive Pulmonary Disease: A Systematic Review and Meta-Analysis. Int J Chron Obstruct Pulmon Dis 2023; 18:1681-1690. [PMID: 37551391 PMCID: PMC10404405 DOI: 10.2147/copd.s412626] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/24/2023] [Indexed: 08/09/2023] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disease characterized by neutrophils airway infiltration. It is currently known that Interleukin-17 (IL-17) is an important pro-inflammatory factor. It can promote the accumulation of neutrophils and participate in the chronic inflammatory process of COPD. However, the value of IL-17 levels in the diagnosis and assessment of COPD remains controversial. In view of this, we conducted a systematic review and meta-analysis to assess its relevance. Methods We searched databases such as PubMed, Web of Science, Cochrane Library and Embase to extract original research. Results A total of 10 studies with 2268 participants were included in this meta-analysis. The results showed that the level of serum IL-17 in patients with stable COPD was significantly higher than that in healthy controls (standard mean difference SMD, 1.59, 95% CI 0.84-2.34; p<0.001). Compared with the stable COPD group, the serum IL-17 level in acute exacerbation (AECOPD) was significantly higher (SMD, 1.78, 95% CI 1.22-2.33; p<0.001). The level of IL-17 in sputum of COPD patients was also higher than that of healthy controls (SMD, 2.03, 95% CI 0.74-3.31; p<0.001). Conclusion Our results showed that IL-17 levels were elevated in serum and sputum in COPD patients compared with healthy controls, and IL-17 levels increased with disease progression. IL-17 serves as a potential biomarker to indicate the persistence of neutrophilic inflammation and exacerbation of COPD.
Collapse
Affiliation(s)
- Ru Ma
- The First Clinical Medical College of Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Gansu Provincial People’s Hospital, Lanzhou, Gansu Province, People’s Republic of China
| | - Hongling Su
- The First Clinical Medical College of Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Gansu Provincial People’s Hospital, Lanzhou, Gansu Province, People’s Republic of China
| | - Keping Jiao
- The First Clinical Medical College of Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Gansu Provincial People’s Hospital, Lanzhou, Gansu Province, People’s Republic of China
| | - Jian Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| |
Collapse
|
40
|
Racanelli AC, Choi AMK. CEACAM6: A Novel Marker of Chronic Obstructive Pulmonary Disease Susceptibility? Am J Respir Crit Care Med 2023; 207:1546-1548. [PMID: 37219336 PMCID: PMC10273108 DOI: 10.1164/rccm.202303-0610ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023] Open
Affiliation(s)
- Alexandra C Racanelli
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I Weill Department of Medicine, Weill Cornell Medicine New York, New York, USA and New York Presbyterian Hospital-Weill Cornell Medical Center New York, New York, USA
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I Weill Department of Medicine, Weill Cornell Medicine New York, New York, USA and New York Presbyterian Hospital-Weill Cornell Medical Center New York, New York, USA
| |
Collapse
|
41
|
Wu CY, Cilic A, Pak O, Dartsch RC, Wilhelm J, Wujak M, Lo K, Brosien M, Zhang R, Alkoudmani I, Witte B, Pedersen F, Watz H, Voswinckel R, Günther A, Ghofrani HA, Brandes RP, Schermuly RT, Grimminger F, Seeger W, Sommer N, Weissmann N, Hadzic S. CEACAM6 as a Novel Therapeutic Target to Boost HO-1-mediated Antioxidant Defense in COPD. Am J Respir Crit Care Med 2023; 207:1576-1590. [PMID: 37219322 DOI: 10.1164/rccm.202208-1603oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 05/23/2023] [Indexed: 05/24/2023] Open
Abstract
Rationale: Tobacco smoking and air pollution are primary causes of chronic obstructive pulmonary disease (COPD). However, only a minority of smokers develop COPD. The mechanisms underlying the defense against nitrosative/oxidative stress in nonsusceptible smokers to COPD remain largely unresolved. Objectives: To investigate the defense mechanisms against nitrosative/oxidative stress that possibly prevent COPD development or progression. Methods: Four cohorts were investigated: 1) sputum samples (healthy, n = 4; COPD, n = 37), 2) lung tissue samples (healthy, n = 13; smokers without COPD, n = 10; smoker+COPD, n = 17), 3) pulmonary lobectomy tissue samples (no/mild emphysema, n = 6), and 4) blood samples (healthy, n = 6; COPD, n = 18). We screened 3-nitrotyrosine (3-NT) levels, as indication of nitrosative/oxidative stress, in human samples. We established a novel in vitro model of a cigarette smoke extract (CSE)-resistant cell line and studied 3-NT formation, antioxidant capacity, and transcriptomic profiles. Results were validated in lung tissue, isolated primary cells, and an ex vivo model using adeno-associated virus-mediated gene transduction and human precision-cut lung slices. Measurements and Main Results: 3-NT levels correlate with COPD severity of patients. In CSE-resistant cells, nitrosative/oxidative stress upon CSE treatment was attenuated, paralleled by profound upregulation of heme oxygenase-1 (HO-1). We identified carcinoembryonic antigen cell adhesion molecule 6 (CEACAM6) as a negative regulator of HO-1-mediated nitrosative/oxidative stress defense in human alveolar type 2 epithelial cells (hAEC2s). Consistently, inhibition of HO-1 activity in hAEC2s increased the susceptibility toward CSE-induced damage. Epithelium-specific CEACAM6 overexpression increased nitrosative/oxidative stress and cell death in human precision-cut lung slices on CSE treatment. Conclusions: CEACAM6 expression determines the hAEC2 sensitivity to nitrosative/oxidative stress triggering emphysema development/progression in susceptible smokers.
Collapse
Affiliation(s)
- Cheng-Yu Wu
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
| | - Anis Cilic
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
| | - Oleg Pak
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
| | - Ruth Charlotte Dartsch
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
| | - Jochen Wilhelm
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University Giessen, Giessen, Germany
| | - Magdalena Wujak
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
- Department of Medicinal Chemistry, Collegium Medicum in Bydgoszcz, Faculty of Pharmacy, Nicolaus Copernicus University in Torun, Bydgoszcz, Poland
| | - Kevin Lo
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
| | - Monika Brosien
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
| | - Ruoyu Zhang
- Department of General and Thoracic Surgery, University Hospital of Giessen, Giessen, Germany
| | - Ibrahim Alkoudmani
- Department of General and Thoracic Surgery, University Hospital of Giessen, Giessen, Germany
| | - Biruta Witte
- Department of General and Thoracic Surgery, University Hospital of Giessen, Giessen, Germany
| | - Frauke Pedersen
- Pulmonary Research Institute at LungenClinic Grosshansdorf, Airway Research Center North, DZL, Grosshansdorf, Germany
| | - Henrik Watz
- Pulmonary Research Institute at LungenClinic Grosshansdorf, Airway Research Center North, DZL, Grosshansdorf, Germany
| | | | - Andreas Günther
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
| | - Hossein A Ghofrani
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany; and
| | - Ralph T Schermuly
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
| | - Friedrich Grimminger
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University Giessen, Giessen, Germany
| | - Werner Seeger
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
- Institute for Lung Health (ILH), Justus Liebig University Giessen, Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Natascha Sommer
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
| | - Stefan Hadzic
- Excellence Cluster Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, member of the German Center for Lung Research (DZL), Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
42
|
Baranasic J, Niazi Y, Chattopadhyay S, Rumora L, Ćorak L, Dugac AV, Jakopović M, Samaržija M, Försti A, Knežević J. Germline variants of the genes involved in NF-kB activation are associated with the risk of COPD and lung cancer development. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2023; 73:243-256. [PMID: 37307368 DOI: 10.2478/acph-2023-0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/09/2023] [Indexed: 06/14/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer (LC) are closely related diseases associated with smoking history and dysregulated immune response. However, not all smokers develop the disease, indicating that genetic susceptibility could be important. Therefore, the aim of this study was to search for the potential overlapping genetic biomarkers, with a focus on single nucleotide polymorphisms (SNPs) located in the regulatory regions of immune-related genes. Additionally, the aim was to see if an identified SNP has potentially an effect on proinflamma-tory cytokine concentration in the serum of COPD patients. We extracted summary data of variants in 1511 immune-related genes from COPD and LC genome-wide association studies (GWAS) from the UK Biobank. The LC data had 203 cases, patients diagnosed with LC, and 360 938 controls, while COPD data had 1 897 cases and 359 297 controls. Assuming 1 association/gene, SNPs with a p-value < 3.3 × 10-5 were considered statistically significantly associated with the disease. We identified seven SNPs located in different genes (BAG6, BTNL2, TNF, HCP5, MICB, NCR3, ABCF1, TCF7L1) to be associated with the COPD risk and two with the LC risk (HLA-C, HLA-B), with statistical significance. We also identified two SNPs located in the IL2RA gene associated with LC (rs2386841; p = 1.86 × 10-4) and COPD (rs11256442; p = 9.79 × 10-3) but with lower significance. Functional studies conducted on COPD patients showed that RNA expression of IL2RA, IFNγ and related proinflammatory cytokines in blood serum did not correlate with a specific genotype. Although results presented in this study do not fully support our hypothesis, it is worth to mention that the identified genes/SNPs that were associated with either COPD or LC risk, all were involved in the activation of the NF-κB transcription factor which is closely related to the regulation of the inflammatory response, a condition associated with both pathologies.
Collapse
Affiliation(s)
- Jurica Baranasic
- 1Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia
| | - Yasmeen Niazi
- 2Hopp Children's Cancer Center (KiTZ) Heidelberg, Germany
- 3Division of Pediatric Neurooncology German Cancer Research Center (DKFZ) German Cancer Consortium (DKTK) Heidelberg, Germany
| | - Subhayan Chattopadhyay
- 3Division of Pediatric Neurooncology German Cancer Research Center (DKFZ) German Cancer Consortium (DKTK) Heidelberg, Germany
- 4Departments of Clinical Genetics, Lund University, Lund, Sweden
| | - Lada Rumora
- 5Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb Zagreb, Croatia
| | - Lorna Ćorak
- 6Clinical Department for Respiratory Diseases Jordanovac, University Hospital Zagreb, School of Medicine University of Zagreb, Zagreb, Croatia
| | - Andrea Vukić Dugac
- 6Clinical Department for Respiratory Diseases Jordanovac, University Hospital Zagreb, School of Medicine University of Zagreb, Zagreb, Croatia
| | - Marko Jakopović
- 6Clinical Department for Respiratory Diseases Jordanovac, University Hospital Zagreb, School of Medicine University of Zagreb, Zagreb, Croatia
| | - Miroslav Samaržija
- 6Clinical Department for Respiratory Diseases Jordanovac, University Hospital Zagreb, School of Medicine University of Zagreb, Zagreb, Croatia
| | - Asta Försti
- 2Hopp Children's Cancer Center (KiTZ) Heidelberg, Germany
- 3Division of Pediatric Neurooncology German Cancer Research Center (DKFZ) German Cancer Consortium (DKTK) Heidelberg, Germany
| | - Jelena Knežević
- 1Division of Molecular Medicine, Rudjer Boskovic Institute, Zagreb, Croatia
- 7Faculty of Dental Medicine and Health University of Osijek, Osijek, Croatia
| |
Collapse
|
43
|
Chen R, Cui Y, Mak JCW. Novel treatments against airway inflammation in COPD based on drug repurposing. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 98:225-247. [PMID: 37524488 DOI: 10.1016/bs.apha.2023.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a major cause of death and reduces quality of life that contributes to a health problem worldwide. Chronic airway inflammation is a hallmark of COPD, which occurs in response to exposure of inhaled irritants like cigarette smoke. Despite accessible to the most up-to-date medications, none of the treatments is currently available to decrease the disease progression. Therefore, it is believed that drugs which can reduce airway inflammation will provide effective disease modifying therapy for COPD. There are many broad-range anti-inflammatory drugs including those that inhibit cell signaling pathways like inhibitors of p38 mitogen-activated protein kinase (MAPK), nuclear factor-κB (NF-κB), and phosphoinositide-3-kinase (PI3K), are now in phase III development for COPD. In this chapter, we review recent basic research data in the laboratory that may indicate novel therapeutic pathways arisen from currently used drugs such as selective monoamine oxidase (MAO)-B inhibitors and drugs targeting peripheral benzodiazepine receptors [also known as translocator protein (TSPO)] to reduce airway inflammation. Considering the impact of chronic airway inflammation on the lives of COPD patients, the potential pharmacological candidates for new anti-inflammatory targets should be further investigated. In addition, it is crucial to consider the phenotypes/molecular endotypes of COPD patients together with specific outcome measures to target novel therapies. This review will enhance our knowledge on how cigarette smoke affects MAO-B activity and TSPO activation/inactivation with specific ligands through regulation of mitochondrial function, and will help to identify new potential treatment for COPD in future.
Collapse
Affiliation(s)
- Rui Chen
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P.R. China; Centre for Immunology and Infection, Hong Kong Science Park, Hong Kong SAR, P.R. China
| | - Yuting Cui
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, P.R. China
| | - Judith C W Mak
- Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P.R. China.
| |
Collapse
|
44
|
Li K, Mei X, Xu K, Jia L, Zhao P, Tian Y, Li J. Comparative study of cigarette smoke, Klebsiella pneumoniae, and their combination on airway epithelial barrier function in mice. ENVIRONMENTAL TOXICOLOGY 2023; 38:1133-1142. [PMID: 36757011 DOI: 10.1002/tox.23753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/09/2023] [Accepted: 01/27/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND The airway epithelium acts as a physical barrier to protect pulmonary airways against pathogenic microorganisms and toxic substances, such as cigarette smoke (CS), bacteria, and viruses. The disruption of the structural integrity and dysfunction of the airway epithelium is related to the occurrence and progression of chronic obstructive pulmonary disease. PURPOSE The aim of this study is to compare the effects of CS, Klebsiella pneumoniae (KP), and their combination on airway epithelial barrier function. METHODS The mice were exposed to CS, KP, and their combination from 1 to 8 weeks. After the cessation of CS and KP at Week 8, we observed the recovery of epithelial barrier function in mice for an additional 16 weeks. To compare the epithelial barrier function among different groups over time, the mice were sacrificed at Weeks 4, 8, 16, and 24 and then the lungs were harvested to detect the pulmonary pathology, inflammatory cytokines, and tight junction proteins. To determine the underlying mechanisms, the BEAS-2B cells were treated with an epidermal growth factor receptor (EGFR) inhibitor (AG1478). RESULTS The results of this study suggested that the decreased lung function, increased bronchial wall thickness (BWT), elevated inflammatory factors, and reduced tight junction protein levels were observed at Week 8 in CS-induced mice and these changes persisted until Week 16. In the KP group, increased BWT and elevated inflammatory factors were observed only at Week 8, whereas in the CS + KP group, decreased lung function, lung tissue injury, inflammatory cell infiltration, and epithelial barrier impairment were observed at Week 4 and persisted until Week 24. To further determine the mechanisms of CS, bacteria, and their combination on epithelial barrier injury, we investigated the changes of EGFR and its downstream protein in the lung tissues of mice and BEAS-2B cells. Our research indicated that CS, KP, or their combination could activate EGFR, which can phosphorylate and activate ERK1/2, and this effect was more pronounced in the CS + KP group. Furthermore, the EGFR inhibitor AG1478 suppressed the phosphorylation of ERK1/2 and subsequently upregulated the expression of ZO-1 and occludin. In general, these results indicated that the combination of CS and KP caused more severe and enduring damage to epithelial barrier function than CS or KP alone, which might be associated with EGFR/ERK1/2 signaling. CONCLUSION Epithelial barrier injury occurred earlier, was more severe, and had a longer duration when induced by the combination of CS and KP compared with the exposure to CS or KP alone, which might be associated with EGFR/ERK signaling.
Collapse
Affiliation(s)
- Kangchen Li
- Department of Respiratory Diseases, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaofeng Mei
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Kexin Xu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lidan Jia
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Peng Zhao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yange Tian
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jiansheng Li
- Department of Respiratory Diseases, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
45
|
Ciminieri C, Woest ME, Reynaert NL, Heijink IH, Wardenaar R, Spierings DCJ, Brandsma CA, Königshoff M, Gosens R. IL-1β Induces a Proinflammatory Fibroblast Microenvironment that Impairs Lung Progenitors' Function. Am J Respir Cell Mol Biol 2023; 68:444-455. [PMID: 36608844 DOI: 10.1165/rcmb.2022-0209oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 01/06/2023] [Indexed: 01/08/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by a persistent inflammatory state in the lungs and defective tissue repair. Although the inflammatory response in patients with COPD is well characterized and known to be exaggerated during exacerbations, its contribution to lung injury and abnormal repair is still unclear. In this study, we aimed to investigate how the inflammatory microenvironment affects the epithelial progenitors and their supporting mesenchymal niche cells involved in tissue repair of the distal lung. We focused on IL-1β, a key inflammatory mediator that is increased during exacerbations of COPD, and used an organoid model of lung epithelial cells and fibroblasts to assess the effect of IL-1β treatment on these cells' transcriptome and secreted factors. Whereas direct treatment of the lung organoids with IL-1β promoted organoid growth, this switched toward inhibition when it was added as fibroblast pretreatment followed by organoid treatment. We then investigated the IL-1β-driven mechanisms in the fibroblasts and found an inflammatory response related to (C-X-C motif) ligand (CXCL) chemokines; we confirmed that these chemokines were responsible for the impaired organoid growth and found that targeting their C-X-C chemokine receptors 1/2 (CXCR1/2) receptors or the IL-1β intracellular signaling reduced the proinflammatory response and restored organoid growth. These data demonstrate that IL-1β alters the fibroblasts' state by promoting a distinct inflammatory response, switching their supportive function on epithelial progenitors toward an inhibitory one in an organoid assay. These results imply that chronic inflammation functions as a shift toward inhibition of repair, thereby contributing to chronic inflammatory diseases like COPD.
Collapse
Affiliation(s)
- Chiara Ciminieri
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD
| | - Manon E Woest
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD
- Aquilo BV, Groningen, The Netherlands
| | - Niki L Reynaert
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht University Medical Center, Maastricht, The Netherlands; and
| | - Irene H Heijink
- Groningen Research Institute for Asthma and COPD
- Groningen Department of Pathology and Medical Biology
- Groningen Department of Pulmonary Diseases, and
| | - René Wardenaar
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Diana C J Spierings
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Corry-Anke Brandsma
- Groningen Research Institute for Asthma and COPD
- Groningen Department of Pathology and Medical Biology
| | - Melanie Königshoff
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD
| |
Collapse
|
46
|
Deskin B. IL-1β Signaling in the Alveolar Niche: Context Is Everything. Am J Respir Cell Mol Biol 2023; 68:356-357. [PMID: 36893326 PMCID: PMC10112426 DOI: 10.1165/rcmb.2023-0008ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023] Open
Affiliation(s)
- Brian Deskin
- The John W. Deming Department of Medicine Tulane University School of Medicine New Orleans, Louisiana
| |
Collapse
|
47
|
Dimic-Janjic S, Hoda MA, Milenkovic B, Kotur-Stevuljevic J, Stjepanovic M, Gompelmann D, Jankovic J, Miljkovic M, Milin-Lazovic J, Djurdjevic N, Maric D, Milivojevic I, Popevic S. The usefulness of MMP-9, TIMP-1 and MMP-9/TIMP-1 ratio for diagnosis and assessment of COPD severity. Eur J Med Res 2023; 28:127. [PMID: 36935521 PMCID: PMC10026402 DOI: 10.1186/s40001-023-01094-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 03/10/2023] [Indexed: 03/21/2023] Open
Abstract
BACKGROUND Inflammation, oxidative stress and an imbalance between proteases and protease inhibitors are recognized pathophysiological features of chronic obstructive pulmonary disease (COPD). The aim of this study was to evaluate serum levels of matrix metalloproteinase-9 (MMP-9) and tissue inhibitor of metalloproteinase-1 (TIMP-1) in patients with COPD and to assess their relationship with lung function, symptom severity scores and recent acute exacerbations. METHODS In this observational cohort study, serum levels of MMP-9 and TIMP-1 and the MMP-9/TIMP-1 ratio in the peripheral blood of COPD patients with stable disease and healthy controls were determined, and their association with lung function (postbronchodilator spirometry, body plethysmography, single breath diffusion capacity for carbon monoxide), symptom severity scores (mMRC and CAT) and exacerbation history were assessed. RESULTS COPD patients (n = 98) had significantly higher levels of serum MMP-9 and TIMP-1 and a higher MMP-9/TIMP-1 ratio than healthy controls (n = 47) (p ≤ 0.001). The areas under the receiver operating characteristic curve for MMP-9, TIMP-1 and the MMP-9/TIMP-1 ratio for COPD diagnosis were 0.974, 0.961 and 0.910, respectively (all p < 0.05). MMP-9 and the MMP-9/TIMP-1 ratio were both negatively correlated with FVC, FEV1, FEV1/FVC, VC, and IC (all p < 0.05). For MMP-9, a positive correlation was found with RV/TLC% (p = 0.005), and a positive correlation was found for the MMP-9/TIMP-1 ratio with RV% and RV/TLC% (p = 0.013 and 0.002, respectively). Patients with COPD GOLD 3 and 4 presented greater MMP-9 levels and a greater MMP-9/TIMP-1 ratio compared to GOLD 1 and 2 patients (p ≤ 0.001). No correlation between diffusion capacity for carbon monoxide and number of acute exacerbations in the previous year was found. CONCLUSIONS COPD patients have elevated serum levels of MMP-9 and TIMP-1 and MMP-9/TIMP-1 ratio. COPD patients have an imbalance between MMP-9 and TIMP-1 in favor of a pro-proteolytic environment, which overall indicates the importance of the MMP-9/TIMP-1 ratio as a potential biomarker for COPD diagnosis and severity.
Collapse
Affiliation(s)
- Sanja Dimic-Janjic
- Faculty of Medicine, University of Belgrade, Clinic for Pulmonology, University Clinical Center of Serbia, Dr Subotica 8, Belgrade, Serbia.
| | - Mir Alireza Hoda
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Branislava Milenkovic
- Faculty of Medicine, University of Belgrade, Clinic for Pulmonology, University Clinical Center of Serbia, Dr Subotica 8, Belgrade, Serbia
| | - Jelena Kotur-Stevuljevic
- Faculty of Pharmacy, Department for Medical Biochemistry, University of Belgrade, Belgrade, Serbia
| | - Mihailo Stjepanovic
- Faculty of Medicine, University of Belgrade, Clinic for Pulmonology, University Clinical Center of Serbia, Dr Subotica 8, Belgrade, Serbia
| | - Daniela Gompelmann
- Division of Pulmonology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Jelena Jankovic
- Faculty of Medicine, University of Belgrade, Clinic for Pulmonology, University Clinical Center of Serbia, Dr Subotica 8, Belgrade, Serbia
| | - Milica Miljkovic
- Faculty of Pharmacy, Department for Medical Biochemistry, University of Belgrade, Belgrade, Serbia
| | - Jelena Milin-Lazovic
- Faculty of Medicine, Institute for Medical Statistics and Informatics, University of Belgrade, Belgrade, Serbia
| | - Natasa Djurdjevic
- Clinic for Pulmonology, University Clinical Center of Serbia, Koste Todorovica 26, Belgrade, Serbia
| | - Dragana Maric
- Faculty of Medicine, University of Belgrade, Clinic for Pulmonology, University Clinical Center of Serbia, Dr Subotica 8, Belgrade, Serbia
| | - Ivan Milivojevic
- Clinic for Pulmonology, University Clinical Center of Serbia, Koste Todorovica 26, Belgrade, Serbia
| | - Spasoje Popevic
- Faculty of Medicine, University of Belgrade, Clinic for Pulmonology, University Clinical Center of Serbia, Dr Subotica 8, Belgrade, Serbia
| |
Collapse
|
48
|
Song S, Ding L, Liu G, Chen T, Zhao M, Li X, Li M, Qi H, Chen J, Wang Z, Wang Y, Ma J, Wang Q, Li X, Wang Z. The protective effects of baicalin for respiratory diseases: an update and future perspectives. Front Pharmacol 2023; 14:1129817. [PMID: 37007037 PMCID: PMC10060540 DOI: 10.3389/fphar.2023.1129817] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/13/2023] [Indexed: 03/17/2023] Open
Abstract
Background: Respiratory diseases are common and frequent diseases. Due to the high pathogenicity and side effects of respiratory diseases, the discovery of new strategies for drug treatment is a hot area of research. Scutellaria baicalensis Georgi (SBG) has been used as a medicinal herb in China for over 2000 years. Baicalin (BA) is a flavonoid active ingredient extracted from SBG that BA has been found to exert various pharmacological effects against respiratory diseases. However, there is no comprehensive review of the mechanism of the effects of BA in treating respiratory diseases. This review aims to summarize the current pharmacokinetics of BA, baicalin-loaded nano-delivery system, and its molecular mechanisms and therapeutical effects for treating respiratory diseases.Method: This review reviewed databases such as PubMed, NCBI, and Web of Science from their inception to 13 December 2022, in which literature was related to “baicalin”, “Scutellaria baicalensis Georgi”, “COVID-19”, “acute lung injury”, “pulmonary arterial hypertension”, “asthma”, “chronic obstructive pulmonary disease”, “pulmonary fibrosis”, “lung cancer”, “pharmacokinetics”, “liposomes”, “nano-emulsions”, “micelles”, “phospholipid complexes”, “solid dispersions”, “inclusion complexes”, and other terms.Result: The pharmacokinetics of BA involves mainly gastrointestinal hydrolysis, the enteroglycoside cycle, multiple metabolic pathways, and excretion in bile and urine. Due to the poor bioavailability and solubility of BA, liposomes, nano-emulsions, micelles, phospholipid complexes, solid dispersions, and inclusion complexes of BA have been developed to improve its bioavailability, lung targeting, and solubility. BA exerts potent effects mainly by mediating upstream oxidative stress, inflammation, apoptosis, and immune response pathways. It regulates are the NF-κB, PI3K/AKT, TGF-β/Smad, Nrf2/HO-1, and ERK/GSK3β pathways.Conclusion: This review presents comprehensive information on BA about pharmacokinetics, baicalin-loaded nano-delivery system, and its therapeutic effects and potential pharmacological mechanisms in respiratory diseases. The available studies suggest that BA has excellent possible treatment of respiratory diseases and is worthy of further investigation and development.
Collapse
Affiliation(s)
- Siyu Song
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Lu Ding
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Guangwen Liu
- GCP Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Tian Chen
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Meiru Zhao
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xueyan Li
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Min Li
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Hongyu Qi
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jinjin Chen
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Ziyuan Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Ying Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jing Ma
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Qi Wang
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xiangyan Li
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
- *Correspondence: Xiangyan Li, ; Zeyu Wang,
| | - Zeyu Wang
- Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Ministry of Education, Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
- *Correspondence: Xiangyan Li, ; Zeyu Wang,
| |
Collapse
|
49
|
Yang N, Zhang L, Tian D, Wang P, Men K, Ge Y, Zhang C. Tanshinone increases Hemopexin expression in lung cells and macrophages to protect against cigarette smoke-induced COPD and enhance antiviral responses. Cell Cycle 2023; 22:645-665. [PMID: 36218263 PMCID: PMC9980497 DOI: 10.1080/15384101.2022.2129933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/01/2022] [Accepted: 09/26/2022] [Indexed: 11/03/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disease, while respiratory infections can elicit exacerbations in COPD patients to mediate increased mortality. Administration of Tanshinones (TS) derivatives has been demonstrated to protect against cigarette smoking (CS) and lipopolysaccharide (LPS)-induced COPD progression. However, the underlying molecular mechanisms and the roles of TS in mitigating the severity of viral-mediated exacerbations of COPD have not been elucidated. Here, we found that TS treatments significantly attenuated lung function decline, inflammatory responses and oxidative stress in CS and LPS-induced COPD mice. Subsequent RNA-seq analysis revealed significantly upregulated Hemopexin expression and enriched interferons (IFNs) signaling pathways in lung tissues of COPD mice upon TS treatments. Moreover, TS administration demonstrated Hemopexin-dependent beneficial roles in BEAS-2B lung cells and RAW264.7 macrophages, which was associated with the suppression of oxidative stress and ERK, NF-κB, and NLRP3 inflammasome signaling pathways-mediated inflammation. Furthermore, TS promoted IFN signaling and rescued impaired antiviral responses in CS and LPS-exposed lung cells that were infected by influenza virus. Notably, hemopexin over-expression in lung cells and macrophages recapitulated the pharmacological activities of TS. Taken together, these results indicate that TS administration is a promising and potential therapeutic strategy for treating COPD and preventing COPD exacerbations.
Collapse
Affiliation(s)
- Ning Yang
- Department of Respiratory, the Affiliated Hospital of Yan’an University, Yan’an, Shaanxi Province, China
| | - Liang Zhang
- Department of Respiratory, the Affiliated Hospital of Yan’an University, Yan’an, Shaanxi Province, China
| | - Dongdong Tian
- Department of Respiratory, the Affiliated Hospital of Yan’an University, Yan’an, Shaanxi Province, China
| | - Ping Wang
- Department of Respiratory, the Affiliated Hospital of Yan’an University, Yan’an, Shaanxi Province, China
| | - Kai Men
- Department of Respiratory, the Affiliated Hospital of Yan’an University, Yan’an, Shaanxi Province, China
| | - Yiliang Ge
- Hengyang Medical School, University of South China, Hunan
| | - Cailian Zhang
- Department of Respiratory, the Affiliated Hospital of Yan’an University, Yan’an, Shaanxi Province, China
| |
Collapse
|
50
|
Healthful and Unhealthful Plant-Based Diets and Chronic Obstructive Pulmonary Disease in U.S. Adults: Prospective Study. Nutrients 2023; 15:nu15030765. [PMID: 36771471 PMCID: PMC9921620 DOI: 10.3390/nu15030765] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/20/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Despite the potential protective effect of a plant-based diet against chronic obstructive pulmonary disease (COPD), it remains unknown whether intake of different types of plant foods is beneficial for COPD. Our aims were to determine whether adherence to the healthful version of a plant-based diet (healthful Plant-based Diet Index (hPDI)) is associated with a lower COPD risk, whereas adherence to the unhealthful version (unhealthful Plant-based Diet Index (uPDI)) is associated with a higher COPD risk. METHODS 46,948 men from the Health Professionals Follow-up Study, 73,592 women from the Nurses' Health Study, and 85,515 women from the Nurses' Health Study II who completed biennial questionnaires from 1984-2018. We derived diet scores from repeated validated food frequency questionnaires. Among 5,661,994 person-years of follow-up, we documented 2605 validated COPD cases between 1984-2018. RESULTS After tight control for smoking and other potential confounders, COPD risk was 46% lower among participants with the highest hPDI score compared to those with the lowest score. Conversely, COPD risk was 39% higher among participants with the highest uPDI. Further adjustment for processed meat intake led to similar results. CONCLUSIONS These findings provide further evidence for consuming a diet that emphasizes healthful plant foods to optimize lung health.
Collapse
|