1
|
Tao L, Zhang Q, Liu L, Wang K, Liu X, Li J, Zhao P. Magnolol preserves the integrity of the intestinal epithelial barrier and mitigates intestinal injury through activation of PPAR γ in COPD rat. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119068. [PMID: 39522848 DOI: 10.1016/j.jep.2024.119068] [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: 09/26/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Magnolia officinalis Rehder & E.H. Wilson is traditionally used in the treatment of gastrointestinal disorders, diarrhea, and cough. Its main active ingredient, magnolol, exhibits protective effects on the lungs and gastrointestinal tract, including the inhibition of inflammation in these organs. AIM OF THE STUDY This work aims to explore the molecular mechanism by which magnolol suppressed Chronic obstructive pulmonary disease (COPD) intestinal damage by improving the intestinal epithelial barrier. MATERIALS AND METHODS The study focused on investigating the mitigation effect of magnolol on intestinal injury and epithelial barrier in a COPD rat. Caco-2 cells were induced with TNF-α or IL-1β to establish the barrier injury model in order to explore the direct protective effect of magnolol on the intestinal barrier and elucidate the molecular mechanism by which it activates peroxisome proliferators-activated receptors-γ (PPARγ). RESULTS Magnolol significantly improves pulmonary function and tissue damage in COPD rats by inhibiting inflammation, protease imbalance, and oxidative stress. It also suppresses colon tissue damage and inflammation, and protects colon epithelial barrier function by suppressing the decline of tight junction proteins, reducing colon epithelial permeability. In Caco-2 cells, magnolol directly reduces monolayer permeability, increases TEER, and upregulates tight junction protein expression induced by TNF-α or IL-1β. Drug Affinity Responsive Target Stability (DARTS) and thermal shift assays show that magnolol effectively binds to SRC, activating PPARγ signaling in Caco-2 cells and colon tissues of COPD rats. Furthermore, magnolol enhances the binding of PPARγ and RXRα, promoting their activation and entry into the nucleus. The PPARγ inhibitor GW9662 can reverse the effects of magnolol on PPARγ activation and tight junction protein upregulation in IL-1β or TNF-α induced Caco-2 cells. CONCLUSIONS This work demonstrates that magnolol enhances lung and intestinal functions in COPD rats, and elucidates its mechanism of action in protecting the intestinal epithelial barrier by activating PPARγ.
Collapse
Affiliation(s)
- Liuying Tao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, 450046, Henan Province, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Qin Zhang
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, 450046, Henan Province, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Lan Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, 450046, Henan Province, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Kun Wang
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, 450046, Henan Province, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Xuefang Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, 450046, Henan Province, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, 450046, Henan Province, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450000, China
| | - Peng Zhao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, 450046, Henan Province, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province & Education Ministry of PR China, Zhengzhou, 450046, Henan Province, China; Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450000, China.
| |
Collapse
|
2
|
Park JH. Clinical Characteristics of Chronic Obstructive Pulmonary Disease according to Smoking Status. Tuberc Respir Dis (Seoul) 2025; 88:14-25. [PMID: 39474731 DOI: 10.4046/trd.2024.0060] [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: 04/18/2024] [Accepted: 10/28/2024] [Indexed: 01/07/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) can be caused by various factors, including lung infections, asthma, air pollution, childhood growth disorders, and genetic factors, though smoking is the predominant risk factor. The main pathological mechanisms in COPD involve small airway disease, emphysema, mucus hypersecretion, and vascular disorders. COPD in non-smokers is characterized by a normal 1-second forced expiratory volume decline, equal sex distribution, younger age of onset, fewer comorbidities, milder airflow obstruction, preserved diffusing capacity of the lungs for carbon monoxide, and radiological features such as more air-trapping and less severe emphysema compared to COPD in smokers. Nevertheless, non-smokers with COPD still experience a high prevalence of acute exacerbations, nearly equal to that of smokers with COPD. Moreover, COPD itself is an independent risk factor for developing lung cancer, regardless of smoking status. Given that COPD coexists with numerous comorbidities, effectively managing these comorbidities is crucial, requiring multifaceted efforts for comprehensive treatment.
Collapse
Affiliation(s)
- Joo Hun Park
- Department of Pulmonary and Critical Care Medicine, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
3
|
Fu Y, Liu Y, Zhong C, Heidari AA, Liu L, Yu S, Chen H, Wu P. An enhanced machine learning-based prognostic prediction model for patients with AECOPD on invasive mechanical ventilation. iScience 2024; 27:111230. [PMID: 39640592 PMCID: PMC11617955 DOI: 10.1016/j.isci.2024.111230] [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: 07/26/2024] [Revised: 09/16/2024] [Accepted: 10/21/2024] [Indexed: 12/07/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) causes irreversible airflow limitations, increasing global morbidity and mortality. Acute exacerbations (AECOPDs) worsen symptoms and may require mechanical ventilation, leading to complications. Understanding factors affecting AECOPD prognosis during mechanical ventilation is crucial. Inspired by rime ice physics, the RIME algorithm has been proposed but it had limitations in feature selection and solution space exploration. We improve RIME by adding a dispersed foraging mechanism and differential crossover operator, creating DDRIME. Our study analyzes patient data to identify factors related to invasive mechanical ventilation in AECOPD. DDRIME's performance is tested against RIME on 83 functions and 12 public datasets for feature selection. It outperformed most algorithms, with bDDRIME_KNN showing high accuracy in predicting AECOPD outcomes. Key indicators-chronic heart failure (CHF), D-dimer (D-D), fungal infection (FI), and pectoral muscle area (PMA)-predicted prognosis with >0.98 accuracy. bDDRIME is thus a valuable tool for predicting AECOPD patients' outcomes on mechanical ventilation.
Collapse
Affiliation(s)
- Yujie Fu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yining Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Chuyue Zhong
- The First Clinical College, Wenzhou Medical University, Wenzhou 325000, China
| | - Ali Asghar Heidari
- School of Surveying and Geospatial Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Lei Liu
- College of Computer Science, Sichuan University, Chengdu, Sichuan 610065, China
| | - Sudan Yu
- Department of Artificial Intelligence, Wenzhou Polytechnic, Wenzhou 325035, China
| | - Huiling Chen
- Department of Computer Science and Artificial Intelligence, Wenzhou University, Wenzhou 325035, China
| | - Peiliang Wu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
4
|
Shi Z, Shao J, Dong C, Song G, Hu Y, Niu Q, Yan Y. Burden of chronic obstructive pulmonary disease attributable to non-optimal temperature, 1990-2044: six countries on the same isotherm. BMC Public Health 2024; 24:3407. [PMID: 39695480 DOI: 10.1186/s12889-024-20622-6] [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: 05/26/2024] [Accepted: 11/05/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND With the frequent occurrence of extreme weather worldwide, non-optimal temperature increased the risk of death from respiratory diseases. The burden of non-optimal temperature on chronic obstructive pulmonary disease (COPD) was quantitatively assessed, and its influencing factors were discussed to provide a basis for the prevention and treatment of COPD. METHODS Based on GBD 2019, we characterized the age-standardized mortality rate (ASMR) and years of life lost rate (ASYR) of COPD attributable to non-optimal temperature in three groups of countries at different isotherms (China and USA, South Africa and Australia, Iraq and Portugal) between 1990 and 2019. We constructed the age-period-cohort model to analyze age, period and cohort effects on mortality and the Bayesian age-period-cohort model to predict ASMR in six countries. We analyzed the relationship of socio-demographic index (SDI) with ASMR and ASYR by restricted cubic spline and quantile regression using data from 21 GBD regions. RESULTS ASMR of COPD attributable to non-optimal temperature in 2019 was 11.03/100,000 (China), 5.62/100,000 (USA), 2/100,000 (Australia), 0.93/100,000 (Iraq), 3.74/100,000 (Portugal), 4.13/100,000 (South Africa). Low temperature had a greater impact on COPD. The mortality showed an increasing trend with age, the period effect only showed a decreasing trend in China, and cohort effect showed a decreasing trend. The higher COPD burden was concentrated in areas with SDI values of 0.39-0.78. Implied quantile regression of mortality to SDI fit was meaningful at P5 and P75, and ASYR was at P5, P25, P75, and P95. We predicted an upward trend in COPD ASMR over the next 25 years only in the USA. CONCLUSIONS In COPD burden caused by non-optimal temperature, low temperature played a more important role, and it was affected by sex, age, period, cohort and SDI. Over the next 25 years, ASMR in COPD was predicted to decline in all countries except the USA.
Collapse
Affiliation(s)
- Zhengyang Shi
- Department of Preventive Medicine, School of Medicine, Shihezi University, No. 59, North 2nd Rd, Hong-Shan District, Shihezi, Xinjiang, 832003, China
| | - Jianjiang Shao
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, The Xinjiang Production and Construction Corps, Shihezi, Xinjiang, China
| | - Chenxian Dong
- Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Guanling Song
- Department of Preventive Medicine, School of Medicine, Shihezi University, No. 59, North 2nd Rd, Hong-Shan District, Shihezi, Xinjiang, 832003, China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, The Xinjiang Production and Construction Corps, Shihezi, Xinjiang, China
- Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Yunhua Hu
- Department of Preventive Medicine, School of Medicine, Shihezi University, No. 59, North 2nd Rd, Hong-Shan District, Shihezi, Xinjiang, 832003, China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, The Xinjiang Production and Construction Corps, Shihezi, Xinjiang, China
- Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Qiang Niu
- Department of Preventive Medicine, School of Medicine, Shihezi University, No. 59, North 2nd Rd, Hong-Shan District, Shihezi, Xinjiang, 832003, China
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, The Xinjiang Production and Construction Corps, Shihezi, Xinjiang, China
- Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Yizhong Yan
- Department of Preventive Medicine, School of Medicine, Shihezi University, No. 59, North 2nd Rd, Hong-Shan District, Shihezi, Xinjiang, 832003, China.
- Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, The Xinjiang Production and Construction Corps, Shihezi, Xinjiang, China.
- Key Laboratory of Preventive Medicine, Shihezi University, Shihezi, Xinjiang, China.
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases (Ministry of Education), School of Medicine, Shihezi University, Shihezi, Xinjiang, China.
| |
Collapse
|
5
|
Tao L, Zhang Q, Liu L, Wang K, Wang J, Liu X, Zhao P, Li J. Inhibition of AhR disrupts intestinal epithelial barrier and induces intestinal injury by activating NF-κB in COPD. FASEB J 2024; 38:e70256. [PMID: 39679871 DOI: 10.1096/fj.202402320r] [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: 09/28/2024] [Revised: 11/25/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is frequently associated with intestinal comorbidities. Damage to the intestinal barrier plays a crucial role in these disorders, leading to increased intestinal and systemic inflammation, and thereby promoting the progression of COPD. This study aims to investigate the mechanism of intestinal epithelial barrier damage, focusing on the roles of the Aryl hydrocarbon Receptor (AhR) and NF-κB in COPD-related intestinal damage. A COPD rat model was induced by cigarette smoke and bacterial infection, while Caco-2/HT29 intestinal epithelial cells were treated with TNF-α or IL-1β to assess intestinal disorder and the underlying mechanisms of barrier damage. COPD rats exhibited significant lung function decline, pathological damage, and inflammatory response in lung tissues. Additionally, significant intestinal injury was observed, accompanied by pronounced colonic pathological damage, an enhanced inflammatory response, and intestinal barrier disruption. This was evidenced by decreased expression of apical junction proteins and elevated serum diamine oxidase levels. Pro-inflammatory cytokines TNF-α or IL-1β significantly downregulated the expression of apical junction proteins in Caco-2/HT29 cells, reduced transepithelial electrical resistance of Caco-2 cells, and increased FD-4 permeability. Moreover, TNF-α or IL-1β induction activated NF-κB in Caco-2/HT29 cells, with a similar activation observed in the colonic tissues of COPD rats. The NF-κB inhibitor PDTC suppressed this activation and protected against intestinal epithelial barrier damage. Furthermore, AhR inhibition was observed both in vitro and in vivo. The AhR activator FICZ inhibited NF-κB activation and mitigated intestinal epithelial barrier damage, whereas the AhR inhibitor CH223191 inhibited AhR and exacerbated intestinal epithelial barrier damage by facilitating NF-κB activation. However, the NF-κB inhibitor PDTC did not significantly affect AhR. Additionally, TNF-α/IL-1β inhibited the binding of AhR and p-NF-κB. Consequently, AhR inhibition can downregulate the expression of apical junction proteins, probably through activation of NF-κB signaling leading to intestinal epithelial barrier damage. This study confirmed the presence of lesions in the lungs and intestines of COPD rats, as well as the associated damage to the intestinal epithelial barrier. The inhibition of AhR followed by the activation of NF-κB has been identified as a critical mechanism underlying the injury to the intestinal epithelial barrier.
Collapse
Affiliation(s)
- Liuying Tao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Qin Zhang
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Lan Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Kun Wang
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Juanhui Wang
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xuefang Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Peng Zhao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan province & Education Ministry of P.R. China, Zhengzhou, Henan, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Moscucci F, Bucciarelli V, Gallina S, Sciomer S, Mattioli AV, Maffei S, Nodari S, Pedrinelli R, Andreozzi P, Basili S. Obstructive sleep apnea syndrome (OSAS) in women: A forgotten cardiovascular risk factor. Maturitas 2024; 193:108170. [PMID: 39708590 DOI: 10.1016/j.maturitas.2024.108170] [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: 07/05/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024]
Abstract
Sleep-disordered breathing is a highly prevalent disorder with negative impact on healthcare systems worldwide. This condition has detrimental effects on cardiovascular health and quality of life, and is frequently associated with a variety of comorbidities, including cardiovascular disease, heart failure, diabetes and atrial fibrillation. Nevertheless, it remains frequently undiagnosed and undertreated, especially in specific populations. Studies on sleep-disordered breathing have been conducted mainly on male patients, and the prevalence and severity of this disorder in women are underestimated. Recently, some clinical and laboratory evidence has highlighted the epidemiological and pathophysiological differences between men and women with sleep-disordered breathing. In this review, we discuss sex-related mechanisms of sleep-disordered breathing in frequently associated disorders, to improve clinical understanding of this condition and to simplify the practical application of targeted interventions. The aim is to improve prognosis among female patients and guarantee a better quality of life and a reduction in healthcare costs.
Collapse
Affiliation(s)
- Federica Moscucci
- Azienda Ospedaliera Universitaria Policlinico Umberto I, DAI of Internal Medicine and Medical Specialties, Viale del Policlinico n.155, 00185 Rome, Italy; Department of Clinical Medicine, Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| | - Valentina Bucciarelli
- Cardiovascular Sciences Department, Azienda Ospedaliero-Universitaria delle Marche, 60126 Ancona, Italy
| | - Sabina Gallina
- Department of Neuroscience, Imaging and Clinical Sciences, "G. D'Annunzi" University, Chieti, Italy.
| | - Susanna Sciomer
- Department of Clinical and Internal Medicine, Anesthesiology and Cardiovascular Sciences, University of Rome "Sapienza", Rome, Italy.
| | | | - Silvia Maffei
- Cardiovascular and Gynaecological Endocrinology Unit, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy.
| | - Savina Nodari
- Department of Cardiology, University of Brescia and ASST "Spedali Civili" Hospital, 25123 Brescia, Italy.
| | - Roberto Pedrinelli
- Cardiac, Thoracic and Vascular Department, University of Pisa, 56126 Pisa, Italy.
| | - Paola Andreozzi
- Predictive Medicine Gender Specificity and Chronicity Unit, Department of Internal Medicine, Endocrine-Metabolic Sciences and Infectious Diseases, Azienda Ospedaliero Universitaria Policlinico Umberto I, SIMI National Directive, Rome, Italy.
| | - Stefania Basili
- Department of Translational and Precision Medicine, Sapienza-University of Rome, Viale del Policlinico 155, Rome 00161, Italy.
| |
Collapse
|
7
|
Su J, Qu Y, Wei Y, Wang S, Zhou J, Gao B, Niu C, Zhang Z, Feng W. Effects of core training based on respiratory therapy on elderly stable COPD patients in the rehabilitation hospital: a study protocol for a randomised controlled trial. BMJ Open 2024; 14:e086837. [PMID: 39653565 PMCID: PMC11629001 DOI: 10.1136/bmjopen-2024-086837] [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: 03/24/2024] [Accepted: 11/06/2024] [Indexed: 12/12/2024] Open
Abstract
INTRODUCTION Elderly stable chronic obstructive pulmonary disease (COPD) patients frequently experience walking dysfunction. Research indicates that balance training holds promise for enhancing walking stability in these individuals, whereas respiratory therapy (RT) can enhance walking endurance effectively. However, existing balance training regimens tailored for COPD are intricate and lack specificity, and the impact of combined RT on patients' walking function remains uncertain. This study aims to assess the influence of core training (CT) integrated with RT on walking function among elderly stable COPD patients. METHODS AND ANALYSIS This randomised, assessment-blinded, routine rehabilitation-controlled trial will be carried out at the Department of Respiratory and Critical Care Rehabilitation, the Second Rehabilitation Hospital of Shanghai. A total of 42 elderly patients diagnosed with stable COPD will be randomly allocated to either the study group (SG) or the control group in a 1:1 ratio. Both groups will undergo 6 weeks of standard rehabilitation training. Additionally, patients in the SG will receive CT based on RT. The primary outcome of the study is the 6 min walk test. Secondary outcomes encompass ultrasound indicators of the diaphragm and multifidus, pulmonary function tests, Berg balance scale test, trunk impairment scale test, COPD assessment test and St. George's Respiratory Questionnaire. ETHICS AND DISSEMINATION Ethical approval was obtained from the Ethics Committee of the Second Rehabilitation Hospital of Shanghai (2023-01-01, see online supplemental file 1). All patients will provide written informed consent before participation. The results of the trial are intended for publication in a peer-reviewed journal. TRIAL REGISTRATION NUMBER ChiCTR2400080276.
Collapse
Affiliation(s)
- Jianqing Su
- Department of Pulmonary Rehabilitation, the Second Rehabilitation Hospital of Shanghai, Shanghai, China
- school of rehabilitation science, Shanghai University of Traditional Chinese Medicine, Shanghai, Shanghai, China
| | - Yan Qu
- Department of Pulmonary Rehabilitation, the Second Rehabilitation Hospital of Shanghai, Shanghai, China
| | - Yingjun Wei
- Department of Pulmonary Rehabilitation, the Second Rehabilitation Hospital of Shanghai, Shanghai, China
| | - Shanshan Wang
- Department of Pulmonary Rehabilitation, the Second Rehabilitation Hospital of Shanghai, Shanghai, China
| | - Jingjing Zhou
- Department of Pulmonary Rehabilitation, the Second Rehabilitation Hospital of Shanghai, Shanghai, China
| | - Bingyang Gao
- Department of Pulmonary Rehabilitation, the Second Rehabilitation Hospital of Shanghai, Shanghai, China
| | - Chunlai Niu
- Department of Pulmonary Rehabilitation, the Second Rehabilitation Hospital of Shanghai, Shanghai, China
| | - Zinan Zhang
- Department of Pulmonary Rehabilitation, the Second Rehabilitation Hospital of Shanghai, Shanghai, China
| | - Wei Feng
- Department of Pulmonary Rehabilitation, the Second Rehabilitation Hospital of Shanghai, Shanghai, China
- school of rehabilitation science, Shanghai University of Traditional Chinese Medicine, Shanghai, Shanghai, China
| |
Collapse
|
8
|
Cao K, Miao X, Chen X. Association of inflammation and nutrition-based indicators with chronic obstructive pulmonary disease and mortality. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2024; 43:209. [PMID: 39643902 PMCID: PMC11622568 DOI: 10.1186/s41043-024-00709-x] [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: 08/12/2024] [Accepted: 11/29/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Inflammation and nutrition are strongly linked to respiratory diseases, but the link between inflammation and nutrition-based indicators and chronic obstructive pulmonary disease (COPD) and its mortality has not been reported. METHODS We recruited adults no younger than 20 years old from the NHANES 1999-2018. Inflammation and nutrition-based indicators included NAR, PNI, MAR, RAR, HALP, and ALI. COPD were assessed through a self-report questionnaire. Participants' mortality rates were determined by association with the National Death Index. RESULTS A total of 46,572 individuals were collected in this study, including 1,549 COPD patients. NAR, MAR, and RAR were positively linked with the prevalence of COPD. However, PNI and HALP were negatively linked with the prevalence of COPD. In participants with COPD, the highest quartile of NAR (HR = 1.43 [1.04-1.97]), MAR (HR = 1.66 [1.23-2.26]), and RAR (HR = 2.45 [1.90-3.17]) were linked with an increased risk of all-cause mortality compared to the lowest quartile. However, the highest quartile of PNI (HR = 0.48 [0.38-0.61]) and HALP (HR = 0.56 [0.44-0.71]) were linked with a decreased risk of all-cause mortality compared to the lowest quartile. Randomized survival forests (RSF) showed that RAR had the strongest predictive power for all-cause mortality in COPD individuals among all indicators. CONCLUSION We found that inflammation and nutrition-based indicators were linked to prognosis in COPD patients, with RAR having the highest predictive value.
Collapse
Affiliation(s)
- Kaiqiang Cao
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Ouhai District, Wenzhou, 325000, China
| | - Xiaoyang Miao
- Department of Intensive Care Unit, , the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaorong Chen
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Ouhai District, Wenzhou, 325000, China.
| |
Collapse
|
9
|
Wang D, Yang J, Yang L, Du Y, Zhu Q, Ma C, Zhou D. Combination therapy strategies against multidrug resistant bacteria in vitro and in vivo. Lett Appl Microbiol 2024; 77:ovae129. [PMID: 39674809 DOI: 10.1093/lambio/ovae129] [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: 07/30/2024] [Revised: 11/30/2024] [Accepted: 12/13/2024] [Indexed: 12/16/2024]
Abstract
Exploring effective combination antibacterial therapies has become a research focus. This study selected seven common antibiotics to perform a series of tests on different Gram-negative bacteria isolated from clinical samples of chronic obstructive pulmonary disease patients. More than 70% of the strains exhibited multidrug resistance but remained sensitive to polymyxin B. The checkerboard assay revealed a significant synergistic effect between polymyxin B and tetracycline against different resistant strains, with fractional inhibitory concentration index values consistently below 0.5. Further time-kill curve analysis demonstrated that the use of minimal inhibit concentration of polymyxin B or tetracycline alone had limited bactericidal effects, while their combination significantly reduced bacterial counts by 2-3 log colony-forming units within 12 h. Additionally, the survival rate of larvae treated with the polymyxin B and tetracycline combination was significantly higher than that of the mono-therapy and untreated groups. In brief, this study demonstrates that the combination of polymyxin B and tetracycline exhibits potent antibacterial activity against multidrug resistant Gram-negative bacteria both in vitro and in vivo.
Collapse
Affiliation(s)
- Daliang Wang
- Department of Emergency Medicine, The First People's Hospital Of Jiashan County, Jiaxing 314100 Zhejiang, China
| | - Jie Yang
- Department of Emergency Medicine, The First People's Hospital Of Jiashan County, Jiaxing 314100 Zhejiang, China
| | - Lilan Yang
- Department of Emergency Medicine, The First People's Hospital Of Jiashan County, Jiaxing 314100 Zhejiang, China
| | - Yanglin Du
- Department of Emergency Medicine, The First People's Hospital Of Jiashan County, Jiaxing 314100 Zhejiang, China
| | - Qunchao Zhu
- Department of Emergency Medicine, The First People's Hospital Of Jiashan County, Jiaxing 314100 Zhejiang, China
| | - Chendong Ma
- Department of Emergency Medicine, The First People's Hospital Of Jiashan County, Jiaxing 314100 Zhejiang, China
| | - Dongdong Zhou
- Department of Emergency Medicine, The First People's Hospital Of Jiashan County, Jiaxing 314100 Zhejiang, China
| |
Collapse
|
10
|
Wang Z, Deng M, Xu W, Li C, Zheng Z, Li J, Liao L, Zhang Q, Bian Y, Li R, Miao J, Wang K, Yin Y, Li Y, Zhou X, Hou G. DKK3 as a diagnostic marker and potential therapeutic target for sarcopenia in chronic obstructive pulmonary disease. Redox Biol 2024; 78:103434. [PMID: 39571512 PMCID: PMC11617289 DOI: 10.1016/j.redox.2024.103434] [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: 09/06/2024] [Revised: 10/11/2024] [Accepted: 11/16/2024] [Indexed: 12/08/2024] Open
Abstract
Sarcopenia, characterized by the progressive loss of muscle mass and function, significantly affects patients with chronic obstructive pulmonary disease (COPD) and worsens their morbidity and mortality. The pathogenesis of muscle atrophy in patients with COPD involves complex mechanisms, including protein imbalance and mitochondrial dysfunction, which have been identified in the muscle tissues of patients with COPD. DKK3 (Dickkopf-3) is a secreted glycoprotein involved in the process of myogenesis. However, the role of DKK3 in the regulation of muscle mass is largely unknown. This study investigated the role of DKK3 in COPD-related sarcopenia. DKK3 was found to be overexpressed in cigarette smoking-induced muscle atrophy and in patients with COPD. Importantly, plasma DKK3 levels in COPD patients with sarcopenia were significantly higher than those without sarcopenia, and plasma DKK3 levels could effectively predict sarcopenia in patients with COPD based on two independent cohorts. Mechanistically, DKK3 is secreted by skeletal muscle cells that acts in autocrine and paracrine manners and interacts with the cell surface-activated receptor cytoskeleton-associated protein 4 (CKAP4) to induce mitochondrial dysfunction and myotube atrophy. The inhibition of DKK3 by genetic ablation prevented cigarette smoking-induced skeletal muscle dysfunction. These results suggest that DKK3 is a potential target for the diagnosis and treatment of sarcopenia in patients with COPD.
Collapse
Affiliation(s)
- Zilin Wang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Mingming Deng
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Weidong Xu
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Chang Li
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Ziwen Zheng
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Jiaye Li
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Liwei Liao
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Qin Zhang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Yiding Bian
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Ruixia Li
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Jinrui Miao
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Kai Wang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Yan Yin
- Department of Pulmonary and Critical Care Medicine, First Hospital of China Medical University, Shenyang, China
| | - Yanxia Li
- Respiratory Department, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaoming Zhou
- Department of Pulmonary and Critical Care Medicine, Disease, Fuwai Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Gang Hou
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
11
|
Xu HP, Niu H, Wang H, Lin J, Yao JJ. Knockdown of RTEL1 Alleviates Chronic Obstructive Pulmonary Disease by Modulating M1, M2 Macrophage Polarization and Inflammation. COPD 2024; 21:2316607. [PMID: 38420994 DOI: 10.1080/15412555.2024.2316607] [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: 10/29/2023] [Accepted: 02/05/2024] [Indexed: 03/02/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a common chronic disease characterized by airflow obstruction, which seriously threatens people's health. The COPD mouse model was established with cigarette smoke induction. Hematoxylin-eosin staining and Masson staining were carried out to observe the pathological changes of lung tissues in COPD mice. RTEL1 was silenced in COPD mice, and immunohistochemistry was used to detect RTEL1, ki67 and Caspase-3 expression. The role of RTEL1 in inflammation were evaluated by ELISA, and the impacts of RTEL1 on M1 and M2 macrophage markers (iNOS and CD206) were evaluated by qPCR and western blotting. In COPD model, there was an increase in the number of inflammatory cells, with slightly disorganized cell arrangement, unclear hierarchy, condensed and solidified nuclei, while knockdown of RTEL1 improved the inflammatory infiltration. Moreover, knockdown of RTEL1 reduced ki67-positive cells and increased Caspase-3 positive cells in COPD group. The increased inflammatory factors (IL-1β, MMP-9, TNF-α, IL-4, IL-6, and IL-23) in COPD were suppressed by knockdown of RTEL1, while iNOS was raised and CD206 was inhibited. In conclusion, knockdown of RTEL1 promoted M1 and inhibited M2 macrophage polarization and inflammation to alleviate COPD.
Collapse
Affiliation(s)
- He-Ping Xu
- Department of Emergency Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Huan Niu
- Department of Emergency Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Hong Wang
- Department of Emergency Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Jie Lin
- Department of Emergency Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Jin-Jian Yao
- Department of Emergency Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
12
|
Wada N, Tsunomori A, Kubo T, Hino T, Hata A, Yamada Y, Ueyama M, Nishino M, Kurosaki A, Ishigami K, Kudoh S, Hatabu H. Assessment of pulmonary function in COPD patients using dynamic digital radiography: A novel approach utilizing lung signal intensity changes during forced breathing. Eur J Radiol Open 2024; 13:100579. [PMID: 39041056 PMCID: PMC11260941 DOI: 10.1016/j.ejro.2024.100579] [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: 04/19/2024] [Revised: 05/23/2024] [Accepted: 06/10/2024] [Indexed: 07/24/2024] Open
Abstract
Objectives To investigate the association of lung signal intensity changes during forced breathing using dynamic digital radiography (DDR) with pulmonary function and disease severity in patients with chronic obstructive pulmonary disease (COPD). Methods This retrospective study included 46 healthy subjects and 33 COPD patients who underwent posteroanterior chest DDR examination. We collected raw signal intensity and gray-scale image data. The lung contour was extracted on the gray-scale images using our previously developed automated lung field tracking system and calculated the average of signal intensity values within the extracted lung contour on gray-scale images. Lung signal intensity changes were quantified as SImax/SImin, representing the maximum ratio of the average signal intensity in the inspiratory phase to that in the expiratory phase. We investigated the correlation between SImax/SImin and pulmonary function parameters, and differences in SImax/SImin by disease severity. Results SImax/SImin showed the highest correlation with VC (rs = 0.54, P < 0.0001), followed by FEV1 (rs = 0.44, P < 0.0001), both of which are key indicators of COPD pathophysiology. In a multivariate linear regression analysis adjusted for confounding factors, SImax/SImin was significantly lower in the severe COPD group compared to the normal group (P = 0.0004) and mild COPD group (P=0.0022), suggesting its potential usefulness in assessing COPD severity. Conclusion This study suggests that the signal intensity changes of lung fields during forced breathing using DDR reflect the pathophysiology of COPD and can be a useful index in assessing pulmonary function in COPD patients, potentially improving COPD diagnosis and management.
Collapse
Affiliation(s)
- Noriaki Wada
- Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115, USA
| | - Akinori Tsunomori
- R&D Promotion Division, Healthcare Business Headquarters, KONICA MINOLTA, INC., 2970 Ishikawa-machi, Hachioji-shi, Tokyo 192-8505, Japan
| | - Takeshi Kubo
- Department of Radiology, Tenri Hospital, 200 Mishimacho, Tenri, Nara 632-8552, Japan
| | - Takuya Hino
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Akinori Hata
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshitake Yamada
- Department of Radiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Masako Ueyama
- Department of Health Care, Fukujuji Hospital, Japan Anti-Tuberculosis Association, 3-1-24 Matsuyama, Kiyose, Tokyo 204-8522, Japan
| | - Mizuki Nishino
- Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115, USA
| | - Atsuko Kurosaki
- Department of Diagnostic Radiology, Fukujuji Hospital, Japan Anti-Tuberculosis Association, 3-1-24 Matsuyama, Kiyose, Tokyo 204-8522, Japan
| | - Kousei Ishigami
- Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Shoji Kudoh
- Department of Respiratory Medicine, Fukujuji Hospital, Japan Anti-Tuberculosis Association, 3-1-24 Matsuyama, Kiyose, Tokyo 204-8522, Japan
| | - Hiroto Hatabu
- Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115, USA
| |
Collapse
|
13
|
Xu Y, Li M, Bai L. Pulmonary Epithelium Cell Fate Determination: Chronic Obstructive Pulmonary Disease, Lung Cancer, or Both. Am J Respir Cell Mol Biol 2024; 71:632-645. [PMID: 39078237 DOI: 10.1165/rcmb.2023-0448tr] [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: 12/18/2023] [Accepted: 07/30/2024] [Indexed: 07/31/2024] Open
Abstract
The concurrence of chronic obstructive pulmonary disease (COPD) and lung cancer has been widely reported and extensively addressed by pulmonologists and oncologists. However, most studies have focused on shared risk factors, DNA damage pathways, immune microenvironments, inflammation, and imbalanced proteases/antiproteases. In the present review, we explore the association between COPD and lung cancer in terms of airway pluripotent cell fate determination and discuss the various cell types and signaling pathways involved in the maintenance of lung epithelium homeostasis and their involvement in the pathogenesis of co-occurring COPD and lung cancer.
Collapse
Affiliation(s)
- Yu Xu
- Department of Clinical Oncology, Army Medical Center, and
| | - Mengxia Li
- Department of Clinical Oncology, Army Medical Center, and
| | - Li Bai
- Department of Respiratory and Critical Medicine, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
14
|
Tu Y, Zhang J, Zhao M, He F. Nomogram establishment for short-term survival prediction in ICU patients with aplastic anemia based on the MIMIC-IV database. Hematology 2024; 29:2339778. [PMID: 38625693 DOI: 10.1080/16078454.2024.2339778] [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: 01/24/2024] [Accepted: 03/27/2024] [Indexed: 04/17/2024] Open
Abstract
OBJECTIVE To establish an efficient nomogram model to predict short-term survival in ICU patients with aplastic anemia (AA). METHODS The data of AA patients in the MIMIC-IV database were obtained and randomly assigned to the training set and testing set in a ratio of 7:3. Independent prognosis factors were identified through univariate and multivariate Cox regression analyses. The variance inflation factor was calculated to detect the correlation between variables. A nomogram model was built based on independent prognostic factors and risk scores for factors were generated. Model performance was tested using C-index, receiver operating characteristic (ROC) curve, calibration curve, decision curve analysis (DCA) and Kaplan-Meier curve. RESULTS A total of 1,963 AA patients were included. A nomogram model with 7 variables was built, including SAPS II, chronic pulmonary obstructive disease, body temperature, red cell distribution width, saturation of peripheral oxygen, age and mechanical ventilation. The C-indexes in the training set and testing set were 0.642 and 0.643 respectively, indicating certain accuracy of the model. ROC curve showed favorable classification performance of nomogram. The calibration curve reflected that its probabilistic prediction was reliable. DCA revealed good clinical practicability of the model. Moreover, the Kaplan-Meier curve showed that receiving mechanical ventilation could improve the survival status of AA patients in the short term but did not in the later period. CONCLUSION The nomogram model of the short-term survival rate of AA patients was built based on clinical characteristics, and early mechanical ventilation could help improve the short-term survival rate of patients.
Collapse
Affiliation(s)
- Yan Tu
- Department of Hematology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People's Republic of China
| | - Jingcheng Zhang
- Department of Hematology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People's Republic of China
| | - Mingzhe Zhao
- Department of Hematology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People's Republic of China
| | - Fang He
- Department of Hematology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, People's Republic of China
| |
Collapse
|
15
|
Li L, Feng Q, Yang C. The D-Dimer to Albumin Ratio Could Predict Hospital Readmission Within One Year in Patients with Acute Exacerbation of Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2024; 19:2587-2597. [PMID: 39629180 PMCID: PMC11614582 DOI: 10.2147/copd.s481483] [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: 06/05/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
Purpose To explore the association of D-dimer-to-albumin ratio (DAR) with hospital readmission within one year in patients with acute exacerbation chronic obstructive pulmonary disease (AECOPD). Patients and Methods From January 2019 to October 2022, 509 patients with COPD were enrolled in Baise People's Hospital for this retrospective cohort study. Baseline data and blood samples were collected, and patients were followed up for one year after inclusion. The AECOPD hospital readmission within one year was the outcome. Receiver operating characteristics (ROC) curves were conducted to determine the prognostic performance of DAR for predicting readmission within one year. The relationships between DAR, neutrophil-to-lymphocyte ratio (NLR), and AECOPD hospital readmission were conducted using univariate and multivariate logistic regression models, with odds ratios (ORs) and 95% confidence intervals (CIs). The relationship was further explored in different modified Medical Research Council (mMRC), COPD assessment test (CAT), COPD course, pneumonia, glucocorticoid, antibiotic subgroups. Results Totally, 117 (22.99%) COPD patients were hospital readmission due to AECOPD. The area under the curve (AUC) for the DAR was 0.726. DAR ≥2.21 (OR=1.80, 95% CI: 1.05-3.17) was associated with elevated odds of AECOPD hospital readmission within one year. DAR ≥2.21 was related to increased odds of AECOPD hospital readmission in patients of those mMRC ≥2, CAT >20, COPD course <10 years, and pneumonia. NLR ≥3.69 was associated with higher odds of AECOPD hospital readmission in patients of those mMRC ≥2 and COPD course ≥10 years. Conclusion In patients with AECOPD, DAR showed a better predictive value in predicting the risk of hospital readmission in patients with AECOPD within one year. The findings of our study might help identify patients with a high risk of readmission within one year and provide timely treatment to prevent the reoccurrence of AECOPD.
Collapse
Affiliation(s)
- Li Li
- Department of Respiratory and Critical Care Medicine, Baise People’s Hospital, Baise, Guangxi, 533099, People’s Republic of China
| | - Qinsheng Feng
- Department of Respiratory and Critical Care Medicine, Baise People’s Hospital, Baise, Guangxi, 533099, People’s Republic of China
| | - Chunsong Yang
- Department of Respiratory and Critical Care Medicine, Baise People’s Hospital, Baise, Guangxi, 533099, People’s Republic of China
| |
Collapse
|
16
|
Wang J, Luo Q, Gu T, An F, Zhou Y, Min Y, Zhang R, Jiang Y. Serum-Derived Exosomal TBX2-AS1 Exacerbates COPD by Altering the M1/M2 Ratio of Macrophages through Regulating the miR-423-5p/miR-23b-3p Axis. Immunol Invest 2024:1-25. [PMID: 39589066 DOI: 10.1080/08820139.2024.2434692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
OBJECTIVE To investigate the mechanism of serum exosomes in chronic obstructive pulmonary disease (COPD), especially the effect of lncRNA TBX2-AS1 on macrophage polarization. METHODS Screen differentially expressed genes through bioinformatics analysis, detect the expression of related molecules in clinical samples and cell experiments, construct a mouse model and conduct functional rescue experiments, using various experimental techniques such as RT - qPCR, Western Blot, flow cytometry, ELISA, and luciferase reporter assay. RESULTS TBX2-AS1 is highly expressed in the serum and serum exosomes of COPD patients, and it can promote macrophage M1 polarization and inhibit M2 polarization; it exerts its role by negatively regulating the miR-423-5p/miR-23b - 3p axis, where miR-423-5p inhibits CELSR2 expression to prevent M1 polarization, and miR-23b-3p inhibits NEK6 expression to promote M2 polarization; in vivo experiments, down-regulation of CELSR2/NEK6 can reverse the promoting effect of COPD serum exosomes on lung injury and inflammation. CONCLUSION COPD serum exosomes deliver TBX2-AS1 to macrophages, regulate the miR-423-5p-CELSR2/miR-23b-3p-NEK6 pathway, affect macrophage polarization, and exacerbate the progression of COPD, providing new directions and potential targets for the diagnosis and treatment of COPD.
Collapse
Affiliation(s)
- JinHai Wang
- Department of Emergency, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Qing Luo
- Department of Internal Medicine, People's Hospital of Hainan Tibetan Autonomous Prefecture, Qinghai, Hainan Tibetan Autonomous Prefecture, China
| | - TiJun Gu
- Department of Emergency, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - FenQin An
- Department of Emergency, People's Hospital of Hainan Tibetan Autonomous Prefecture, Hainan Tibetan Autonomous Prefecture, China
| | - YunZheng Zhou
- Department of Emergency, People's Hospital of Hainan Tibetan Autonomous Prefecture, Hainan Tibetan Autonomous Prefecture, China
| | - YePing Min
- Department of Emergency, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - RuiRen Zhang
- Department of Emergency, People's Hospital of Hainan Tibetan Autonomous Prefecture, Hainan Tibetan Autonomous Prefecture, China
| | - YiMing Jiang
- Department of Emergency, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
- Department of Emergency, People's Hospital of Hainan Tibetan Autonomous Prefecture, Hainan Tibetan Autonomous Prefecture, China
| |
Collapse
|
17
|
Liu Y, Li W, Tang J, Gao S. Association of life's essential 8 with chronic obstructive pulmonary disease: a population-based analysis of NHANES 2007-2018. BMC Public Health 2024; 24:3144. [PMID: 39538175 PMCID: PMC11558873 DOI: 10.1186/s12889-024-20534-5] [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: 01/16/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is closely linked to cardiovascular diseases. We aimed to investigate the association between Life's Essential 8 (LE8), the newly established measurement to assess cardiovascular health (CVH), and COPD among U.S. general adults. METHODS This study extracted the National Health and Nutrition Examination Survey (2007-2018) data. Multivariate logistic regression models were used to examine the associations between LE8 and COPD. A restricted cubic spline regression model was used to explore the dose-response relationships between LE8 scores and COPD. In addition, subgroup and sensitivity analyses were performed to assess the robustness of our results. RESULTS Our study included 19,774 participants representing 145.2 million non-institutionalized U.S. population aged ≥ 20 years. The overall age-adjusted prevalence of COPD was 4.5%. After adjusting for the potential covariates, LE8 was inversely associated with COPD [adjusted odds ratio (AOR) = 0.169, 95% CI: 0.115, 0.249], exhibiting a nonlinear dose-response relationship (P for nonlinearity < 0.05). Similar trends in the associations of health behavior score (AOR = 0.300, 95% CI: 0.223, 0.404) and health factor score (AOR = 0.603, 95% CI: 0.426, 0.852) with COPD were also identified. Furthermore, higher LE8 metric scores of nicotine exposure and sleep health were associated with a lower prevalence of COPD. CONCLUSION LE8 was inversely associated with spirometric/self-reported COPD in a nonlinear trend, primarily driven by the nicotine exposure metric of LE8. Adhering to LE8 guidelines, especially smoking cessation, to sustain optimal CVH levels may be beneficial to alleviate the burden of COPD.
Collapse
Affiliation(s)
- Yuhang Liu
- School of Physical Education and Sports, Central China Normal University, Wuhan, 430079, P.R. China
| | - Weidong Li
- School of Physical Education and Sports, Central China Normal University, Wuhan, 430079, P.R. China
| | - Jialing Tang
- Department of Physical Education, Central South University, No. 932 Lushan South Road, Yuelu District, Changsha, 410083, P.R. China
| | - Siyao Gao
- Department of Physical Education, Central South University, No. 932 Lushan South Road, Yuelu District, Changsha, 410083, P.R. China.
| |
Collapse
|
18
|
Guo B, Shi X, Jiang Q, Pan Y, Yang Y, Liu Y, Chen S, Zhu W, Ren L, Liang R, Chen X, Xu H, Wei L, Lin Y, Wang J, Qiu C, Zhou H, Rao L, Wang L, Chen R, Chen S. Targeting Immunoproteasome in Polarized Macrophages Ameliorates Experimental Emphysema Via Activating NRF1/2-P62 Axis and Suppressing IRF4 Transcription. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405318. [PMID: 39356034 PMCID: PMC11600198 DOI: 10.1002/advs.202405318] [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: 05/15/2024] [Revised: 06/22/2024] [Indexed: 10/03/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) stands as the prevailing chronic airway ailment, characterized by chronic bronchitis and emphysema. Current medications fall short in treatment of these diseases, underscoring the urgent need for effective therapy. Prior research indicated immunoproteasome inhibition alleviated various inflammatory diseases by modulating immune cell functions. However, its therapeutic potential in COPD remains largely unexplored. Here, an elevated expression of immunoproteasome subunits LMP2 and LMP7 in the macrophages isolated from mouse with LPS/Elastase-induced emphysema and polarized macrophages in vitro is observed. Subsequently, intranasal administration of the immunoproteasome-specific inhibitor ONX-0914 significantly mitigated COPD-associated airway inflammation and improved lung function in mice by suppressing macrophage polarization. Additionally, ONX-0914 capsulated in PLGA nanoparticles exhibited more pronounced therapeutic effect on COPD than naked ONX-0914 by targeting immunoproteasome in polarized macrophages. Mechanistically, ONX-0914 activated autophagy and endoplasmic reticulum (ER) stress are not attribute to the ONX-0914 mediated suppression of macrophage polarization. Intriguingly, ONX-0914 inhibited M1 polarization through the nuclear factor erythroid 2-related factor-1 (NRF1) and NRF2-P62 axis, while the suppression of M2 polarization is regulated by inhibiting the transcription of interferon regulatory factor 4 (IRF4). In summary, the findings suggest that targeting immunoproteasome in macrophages holds promise as a therapeutic strategy for COPD.
Collapse
Affiliation(s)
- Bingxin Guo
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Xing Shi
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Qiong Jiang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Yuanwei Pan
- Institute of Chemical BiologyShenzhen Bay LaboratoryShenzhen518132China
| | - Yuqiong Yang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, State Key Laboratory of Respiratory DiseaseFirst Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Yuanyuan Liu
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Post‐doctoral Scientific Research Station of Basic Medicine, The Second Clinical Medical CollegeJinan UniversityGuangzhou510632China
| | - Shuyu Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Wenjiao Zhu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Laibin Ren
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Ruifang Liang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
- Department of Respiratory Diseases and Critic Care Unit, Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Post‐doctoral Scientific Research Station of Basic Medicine, The Second Clinical Medical CollegeJinan UniversityGuangzhou510632China
| | - Xue Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Haizhao Xu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Laiyou Wei
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Yongjian Lin
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
- College of PharmacyJinan UniversityGuangzhouGuangdong510632China
| | - Jinyong Wang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Chen Qiu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Haibo Zhou
- College of PharmacyJinan UniversityGuangzhouGuangdong510632China
| | - Lang Rao
- Institute of Chemical BiologyShenzhen Bay LaboratoryShenzhen518132China
| | - Lingwei Wang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Rongchang Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| | - Shanze Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory DiseasesThe First Affiliated Hospital (Shenzhen People's Hospital) and School of Medicine, Southern University of Science and TechnologyShenzhen518055China
| |
Collapse
|
19
|
Li H, Dai X, Zhou J, Wang Y, Zhang S, Guo J, Shen L, Yan H, Jiang H. Mitochondrial dynamics in pulmonary disease: Implications for the potential therapeutics. J Cell Physiol 2024; 239:e31370. [PMID: 38988059 DOI: 10.1002/jcp.31370] [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: 02/26/2024] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
Mitochondria are dynamic organelles that continuously undergo fusion/fission to maintain normal cell physiological activities and energy metabolism. When mitochondrial dynamics is unbalanced, mitochondrial homeostasis is broken, thus damaging mitochondrial function. Accumulating evidence demonstrates that impairment in mitochondrial dynamics leads to lung tissue injury and pulmonary disease progression in a variety of disease models, including inflammatory responses, apoptosis, and barrier breakdown, and that the role of mitochondrial dynamics varies among pulmonary diseases. These findings suggest that modulation of mitochondrial dynamics may be considered as a valid therapeutic strategy in pulmonary diseases. In this review, we discuss the current evidence on the role of mitochondrial dynamics in pulmonary diseases, with a particular focus on its underlying mechanisms in the development of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary disease (COPD), asthma, pulmonary fibrosis (PF), pulmonary arterial hypertension (PAH), lung cancer and bronchopulmonary dysplasia (BPD), and outline effective drugs targeting mitochondrial dynamics-related proteins, highlighting the great potential of targeting mitochondrial dynamics in the treatment of pulmonary disease.
Collapse
Affiliation(s)
- Hui Li
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xinyan Dai
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Junfu Zhou
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yujuan Wang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Shiying Zhang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jiacheng Guo
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Lidu Shen
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Hengxiu Yan
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Huiling Jiang
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| |
Collapse
|
20
|
Fei J, Liu L, Li JF, Zhou Q, Wei Y, Zhou TD, Fu L. Associations of Vitamin D With GPX4 and Iron Parameters in Chronic Obstructive Pulmonary Disease Patients: A Case-Control Study. Can Respir J 2024; 2024:4505905. [PMID: 39502871 PMCID: PMC11535414 DOI: 10.1155/2024/4505905] [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: 10/26/2023] [Revised: 09/10/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Background: Vitamin D deficiency elevates the risk of chronic obstructive pulmonary disease (COPD) patients. Iron parameters elevation and glutathione peroxidase 4 (GPX4) reduction are involved in the process of COPD. The goal is to explore the associations of vitamin D with GPX4 and iron parameters in COPD patients through a case-control study. Methods: COPD patients and control subjects were enrolled. Serum samples and lung tissues were collected. Serum vitamin D and iron levels and pulmonary ferritin and GPX4 expressions were determined. In addition, human pulmonary epithelial cells (BEAS-2B) were incubated with 1,25(OH)2D3 (100 nM), the active form of vitamin D3. Then, vitamin D receptor (VDR) and nuclear factor (erythroid-derived 2)-like 2 (Nrf-2) signaling were detected. Results: In patients with COPD, serum 25-hydroxyvitamin D (25(OH)D) decreased, and iron and ferritin levels in serum and lung tissues increased. Furthermore, pulmonary expression of GPX4 was reduced. Correlative analyzes indicated that lung function was inversely correlated with iron parameters and positively correlated with GPX4. The results showed that serum 25(OH)D deficiency was associated with an elevation in serum iron parameters and a reduction in pulmonary GPX4. In addition, VDR- and Nrf-2-positive lung nuclei were decreased in COPD patients than in control subjects. In patients with COPD, the results indicated a positive relationship between VDR and Nrf-2. Further analysis revealed that Nrf-2-positive nuclei were negatively correlated with iron parameters. In vitro experiments found that 1,25(OH)2D3 treatment activated VDR signaling and elevated the expression of Nrf-2 and GPX4 in BEAS-2B cells. Conclusions: Vitamin D deficiency is positively associated with GPX4 reduction and iron parameters elevation in COPD patients. It is recommended to explore the role of vitamin D supplementation in the progression of COPD.
Collapse
Affiliation(s)
- Jun Fei
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, China
- Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, China
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Bengbu Medical College, Bengbu 233004, Anhui, China
| | - Ling Liu
- Department of Respiratory and Critical Care Medicine, People's Hospital of Yingshan, Fuyang 236000, Anhui, China
| | - Jia-Fei Li
- Department of Respiratory and Critical Care Medicine, First People's Hospital of Chuzhou, Chuzhou 239001, Anhui, China
| | - Qiang Zhou
- Department of Clinical Laboratory, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Yu Wei
- Department of Clinical Laboratory, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Ting-Dong Zhou
- Department of Clinical Laboratory, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Lin Fu
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Bengbu Medical College, Bengbu 233004, Anhui, China
- Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China
| |
Collapse
|
21
|
Alswat AS. The Influence of the Gut Microbiota on Host Health: A Focus on the Gut-Lung Axis and Therapeutic Approaches. Life (Basel) 2024; 14:1279. [PMID: 39459579 PMCID: PMC11509314 DOI: 10.3390/life14101279] [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: 07/29/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
The human gut microbiota is a complex ecosystem harboring thousands of microbial strains that play a crucial role in maintaining the overall well-being of its host. The composition of the gut microbiota varies with age from infancy to adulthood and is influenced by dietary habits, environment, and genetic disposition. Recent advances in culture-independent techniques and nucleic acid sequencing have improved our understanding of the diversity of the gut microbiota. The microbial species present in the gut release short-chain fatty acids (SCFAs), which have anti-inflammatory properties. The gut microbiota also plays a substantial role in modulating the host's immune system, promoting immune tolerance, and maintaining homeostasis. The impact of the gut microbiota on the health of the host is quite evident, as gut dysbiosis has been linked to various diseases, including metabolic disorders, autoimmune diseases, allergies, and inflammatory bowel diseases. The gut microbiota has bidirectional communication with the respiratory system, creating the gut-lung axis, which has been associated with different respiratory diseases. Therapeutic approaches targeting the gut microbiota, such as probiotics, prebiotics, dietary interventions, and fecal microbiota transplantation (FMT), aim to restore microbial balance and promote the growth of beneficial strains in the gut. Nonetheless, gaining knowledge of the complex interactions between the gut microbiota and the host is necessary to develop personalized medicine approaches and microbiota-based therapies for various conditions. This review summarizes studies related to the gut-lung axis with particular emphasis on the role of the microbiota. Future research directions are also discussed.
Collapse
Affiliation(s)
- Amal S Alswat
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
22
|
Cheng H, Wang X, Yao J, Guo N, Liu J. Assessing the causal relationship between non-small cell lung cancer and sepsis: a Mendelian randomization study. BMC Cancer 2024; 24:1233. [PMID: 39375649 PMCID: PMC11457449 DOI: 10.1186/s12885-024-13003-0] [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: 02/28/2024] [Accepted: 09/27/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND A Two-sample Mendelian randomization (MR) Analysis was used to assess the causal relationship between non-small cell lung cancer (NSCLC) and sepsis. METHOD Single nucleotide polymorphisms (SNPs) closely associated with NSCLC were utilized as instrumental variables (IVs) in this study. The Inverse Variance Weighted (IVW) method was used as the primary method for MR analysis, supplemented by the Weighted median, Weighted model, and MR-Egger regression method. Sensitivity analysis was conducted to improve result robustness, and data from various sources were validated and integrated. Bonferroni tests were applied to adjust for multiple comparisons. RESULTS After Bonferroni tests correcting the combined results, MR analysis revealed a significant association between genetically predicted NSCLC and an increased susceptibility to sepsis (odds ratios [OR]: 1.140, 95% confidence interval [CI]: 1.085-1.199, P = 2.61 × 10- 7). The combined results demonstrated that NSCLC is associated with a heightened risk of sepsis in patients under 75 years of age (OR: 1.085, 95%CI: 1.037-1.353, P = 3.84 × 10- 4). Furthermore, lung adenocarcinoma (LUAD) was found to be potentially associated with an increased susceptibility to sepsis (OR: 1.040, 95% CI: 1.009-1.073, P = 1.16 × 10- 2). These results withstood multiple sensitivity analyses, demonstrating their robustness. CONCLUSION This study confirms that NSCLC can significantly increase susceptibility to sepsis at the genetic level, providing valuable insights for the early identification of individuals at risk for sepsis.
Collapse
Affiliation(s)
- Huixin Cheng
- The First Clinical Medical College of Lanzhou University, No. 222 Tianshui South Road, Lanzhou, Gansu Province, 730000, China
| | - Xuehan Wang
- The First Clinical Medical College of Lanzhou University, No. 222 Tianshui South Road, Lanzhou, Gansu Province, 730000, China
| | - Juyi Yao
- Traditional Chinese Medicine Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830039, China
| | - Na Guo
- The First Clinical Medical College of Lanzhou University, No. 222 Tianshui South Road, Lanzhou, Gansu Province, 730000, China
| | - Jian Liu
- The First Clinical Medical College of Lanzhou University, No. 222 Tianshui South Road, Lanzhou, Gansu Province, 730000, China.
- Department of Intensive Care Unit, Gansu Provincial Maternity and Child Health Hospital, Gansu Provincial General Hospital, Lan Zhou, Gansu Province, 730050, China.
| |
Collapse
|
23
|
Chen F, Yang M, Wang H, Liu L, Shen Y, Chen L. High blood eosinophils predict the risk of COPD exacerbation: A systematic review and meta-analysis. PLoS One 2024; 19:e0302318. [PMID: 39361621 PMCID: PMC11449345 DOI: 10.1371/journal.pone.0302318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 04/02/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND The association between blood eosinophils and COPD exacerbation has been controversial. This study aims to investigate whether high blood eosinophils predict the risk of COPD exacerbation across different thresholds and subgroups. METHODS PubMed, Embase and Web of science were searched for randomized controlled trial (RCT) and observational studies regarding the relationship between blood eosinophils and COPD exacerbation. Pooled risk ratio (RR) for COPD exacerbation was calculated using the Mantel-Haenszel method with a random-effects model. RESULTS A total of 21 studies (1 RCT and 20 observational studies) with 79868 participants were included. Thresholds of high blood eosinophils including absolute counts (200, 300 and 400 cell/μL) and percentages (2%, 3% and 4%) were analyzed respectively. Pooled analyses suggested that high blood eosinophils were significantly associated with increased risk of COPD exacerbation when using the thresholds of 300 cells/μL (RR 1.21, 95%CI 1.12-1.30, P <0.001, 16 studies), 400 cells/μL (RR 1.79, 95%CI 1.41-2.28, P <0.001, 3 studies), 2% (RR 1.26, 95%CI 1.02-1.55, P = 0.030, 10 studies) and 4% (RR 1.44, 95%CI 1.05-1.96, P = 0.022, 4 studies), but not 200 cells/μL and 3% (P>0.05). Moreover, high blood eosinophils contributed to moderate-severe exacerbation of COPD by the cutoffs of 300 cells/μL (RR 1.30, 95%CI 1.16-1.45, P<0.001, 11 studies) and 2% (RR 1.33, 95%CI 1.02-1.76, P = 0.037, 8 studies). In subgroup analyses, the pooled results further showed a significant association between high blood eosinophils (especially over 300 cells/μL) and risk of COPD exacerbation among patients from Europe and Asia, and whether with stable or exacerbation phase at baseline, and regardless of the follow-up time (≤ or > 1year). CONCLUSIONS This study demonstrates that high blood eosinophils (over 300 cells/μL or 2%) could predict the risk of moderate-severe exacerbation of COPD in specific subgroups. However, large sample-sized, prospective, and well-designed studies are required to validate the present findings.
Collapse
Affiliation(s)
- Fangying Chen
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Tuberculosis, The Third People’s Hospital of Tibet Autonomous Region, Lhasa, Tibet, China
| | - Mei Yang
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hao Wang
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lian Liu
- Laboratory of Pulmonary Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yongchun Shen
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Chen
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
24
|
Cantor J. The Role of the Extracellular Matrix in the Pathogenesis and Treatment of Pulmonary Emphysema. Int J Mol Sci 2024; 25:10613. [PMID: 39408941 PMCID: PMC11477147 DOI: 10.3390/ijms251910613] [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: 09/02/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Pulmonary emphysema involves progressive destruction of alveolar walls, leading to enlarged air spaces and impaired gas exchange. While the precise mechanisms responsible for these changes remain unclear, there is growing evidence that the extracellular matrix plays a critical role in the process. An essential feature of pulmonary emphysema is damage to the elastic fiber network surrounding the airspaces, which stores the energy needed to expel air from the lungs. The degradation of these fibers disrupts the mechanical forces involved in respiration, resulting in distension and rupture of alveolar walls. While the initial repair process mainly consists of elastin degradation and resynthesis, continued alveolar wall injury may be associated with increased collagen deposition, resulting in a mixed pattern of emphysema and interstitial fibrosis. Due to the critical role of elastic fiber injury in pulmonary emphysema, preventing damage to this matrix component has emerged as a potential therapeutic strategy. One treatment approach involves the intratracheal administration of hyaluronan, a polysaccharide that prevents elastin breakdown by binding to lung elastic fibers. In clinical trials, inhalation of aerosolized HA decreased elastic fiber injury, as measured by the release of the elastin-specific cross-linking amino acids, desmosine, and isodesmosine. By protecting elastic fibers from enzymatic and oxidative damage, aerosolized HA could alter the natural history of pulmonary emphysema, thereby reducing the risk of respiratory failure.
Collapse
Affiliation(s)
- Jerome Cantor
- School of Pharmacy and Allied Health Sciences, St John's University, Queens, NY 11439, USA
| |
Collapse
|
25
|
Hurraß J, Heinzow B, Walser-Reichenbach S, Aurbach U, Becker S, Bellmann R, Bergmann KC, Cornely OA, Engelhart S, Fischer G, Gabrio T, Herr CEW, Joest M, Karagiannidis C, Klimek L, Köberle M, Kolk A, Lichtnecker H, Lob-Corzilius T, Mülleneisen N, Nowak D, Rabe U, Raulf M, Steinmann J, Steiß JO, Stemler J, Umpfenbach U, Valtanen K, Werchan B, Willinger B, Wiesmüller GA. [Medical clinical diagnostics for indoor mould exposure - Update 2023 (AWMF Register No. 161/001)]. Pneumologie 2024; 78:693-784. [PMID: 39424320 DOI: 10.1055/a-2194-6914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
This article is an abridged version of the updated AWMF mould guideline "Medical clinical diagnostics in case of indoor mould exposure - Update 2023", presented in July 2023 by the German Society of Hygiene, Environmental Medicine and Preventive Medicine (Gesellschaft für Hygiene, Umweltmedizin und Präventivmedizin, GHUP), in collaboration with German and Austrian scientific medical societies, and experts. Indoor mould growth is a potential health risk, even if a quantitative and/or causal relationship between the occurrence of individual mould species and health problems has yet to be established. There is no evidence for a causal relationship between moisture/mould damage and human diseases, mainly because of the ubiquitous presence of fungi and hitherto inadequate diagnostic methods. Sufficient evidence for an association between moisture/mould damage and the following health effects has been established for: allergic respiratory diseases, allergic rhinitis, allergic rhino-conjunctivitis, allergic bronchopulmonary aspergillosis (ABPA), other allergic bronchopulmonary mycosis (ABPM), aspergilloma, Aspergillus bronchitis, asthma (manifestation, progression, exacerbation), bronchitis (acute, chronic), community-acquired Aspergillus pneumonia, hypersensitivity pneumonitis (HP; extrinsic allergic alveolitis (EEA)), invasive Aspergillosis, mycoses, organic dust toxic syndrome (ODTS) [workplace exposure], promotion of respiratory infections, pulmonary aspergillosis (subacute, chronic), and rhinosinusitis (acute, chronically invasive, or granulomatous, allergic). In this context the sensitizing potential of moulds is obviously low compared to other environmental allergens. Recent studies show a comparatively low sensitization prevalence of 3-22,5 % in the general population across Europe. Limited or suspected evidence for an association exist with respect to atopic eczema (atopic dermatitis, neurodermatitis; manifestation), chronic obstructive pulmonary disease (COPD), mood disorders, mucous membrane irritation (MMI), odor effects, and sarcoidosis. (iv) Inadequate or insufficient evidence for an association exist for acute idiopathic pulmonary hemorrhage in infants, airborne transmitted mycotoxicosis, arthritis, autoimmune diseases, cancer, chronic fatigue syndrome (CFS), endocrinopathies, gastrointestinal effects, multiple chemical sensitivity (MCS), multiple sclerosis, neuropsychological effects, neurotoxic effects, renal effects, reproductive disorders, rheumatism, sick building syndrome (SBS), sudden infant death syndrome, teratogenicity, thyroid diseases, and urticaria.The risk of infection posed by moulds regularly occurring indoors is low for healthy persons; most species are in risk group 1 and a few in risk group 2 (Aspergillus fumigatus, A. flavus) of the German Biological Agents Act (Biostoffverordnung). Only moulds that are potentially able to form toxins can be triggers of toxic reactions. Whether or not toxin formation occurs in individual cases is determined by environmental and growth conditions, water activity, temperature and above all the growth substrates.In case of indoor moisture/mould damage, everyone can be affected by odor effects and/or mood disorders.However, this is not an acute health hazard. Predisposing factors for odor effects can include genetic and hormonal influences, imprinting, context and adaptation effects. Predisposing factors for mood disorders may include environmental concerns, anxiety, condition, and attribution, as well as various diseases. Risk groups to be protected particularly regarding infection risk are immunocompromised persons according to the classification of the German Commission for Hospital Hygiene and Infection Prevention (Kommission für Krankenhaushygiene und Infektionsprävention, KRINKO) at the Robert Koch-Institute (RKI), persons suffering from severe influenza, persons suffering from severe COVID-19, and persons with cystic fibrosis (mucoviscidosis); with regard to allergic risk, persons with cystic fibrosis (mucoviscidosis) and patients with bronchial asthma must be protected. The rational diagnostics include the medical history, physical examination, and conventional allergy diagnostics including provocation tests if necessary; sometimes cellular test systems are indicated. In the case of mould infections, the reader is referred to the specific guidelines. Regarding mycotoxins, there are currently no useful and validated test procedures for clinical diagnostics. From a preventive medical point of view, it is important that indoor mould infestation in relevant magnitudes cannot be tolerated for precautionary reasons.For evaluation of mould damage in the indoor environment and appropriate remedial procedures, the reader is referred to the mould guideline issued by the German Federal Environment Agency (Umweltbundesamt, UBA).
Collapse
Affiliation(s)
- Julia Hurraß
- Sachgebiet Hygiene in Gesundheitseinrichtungen, Abteilung Infektions- und Umwelthygiene, Gesundheitsamt der Stadt Köln
| | - Birger Heinzow
- Ehemals: Landesamt für soziale Dienste (LAsD) Schleswig-Holstein, Kiel
| | | | - Ute Aurbach
- Labor Dr. Wisplinghoff
- ZfMK - Zentrum für Umwelt, Hygiene und Mykologie, Köln
| | - Sven Becker
- Universitätsklinik für Hals-, Nasen- und Ohrenheilkunde, Universitätsklinikum Tübingen
| | - Romuald Bellmann
- Universitätsklinik für Innere Medizin I, Medizinische Universität Innsbruck
| | | | - Oliver A Cornely
- Translational Research, CECAD Cluster of Excellence, Universität zu Köln
| | | | - Guido Fischer
- Landesgesundheitsamt Baden-Württemberg im Regierungspräsidium Stuttgart
| | - Thomas Gabrio
- Ehemals: Landesgesundheitsamt Baden-Württemberg im Regierungspräsidium Stuttgart
| | - Caroline E W Herr
- Bayerisches Landesamt für Gesundheit und Lebensmittelsicherheit München
- Ludwig-Maximilians-Universität München, apl. Prof. "Hygiene und Umweltmedizin"
| | - Marcus Joest
- Allergologisch-immunologisches Labor, Helios Lungen- und Allergiezentrum Bonn
| | - Christian Karagiannidis
- Fakultät für Gesundheit, Professur für Extrakorporale Lungenersatzverfahren, Universität Witten/Herdecke
- Lungenklinik Köln Merheim, Kliniken der Stadt Köln
| | | | - Martin Köberle
- Klinik und Poliklinik für Dermatologie und Allergologie am Biederstein, Technische Universität München
| | - Annette Kolk
- Institut für Arbeitsschutz der DGUV (IFA), Bereich Biostoffe, Sankt Augustin
| | | | | | | | - Dennis Nowak
- Institut und Poliklinik für Arbeits-, Sozial- und Umweltmedizin, Mitglied Deutsches Zentrum für Lungenforschung, Klinikum der Universität München
| | - Uta Rabe
- Zentrum für Allergologie und Asthma, Johanniter-Krankenhaus Treuenbrietzen
| | - Monika Raulf
- Institut für Prävention und Arbeitsmedizin der Deutschen Gesetzlichen Unfallversicherung, Institut der Ruhr-Universität Bochum (IPA)
| | - Jörg Steinmann
- Institut für Klinikhygiene, Medizinische Mikrobiologie und Klinische Infektiologie, Paracelsus Medizinische Privatuniversität Klinikum Nürnberg
| | - Jens-Oliver Steiß
- Zentrum für Kinderheilkunde und Jugendmedizin, Universitätsklinikum Gießen und Marburg GmbH, Gießen
- Schwerpunktpraxis Allergologie und Kinder-Pneumologie Fulda
| | - Jannik Stemler
- Translational Research, CECAD Cluster of Excellence, Universität zu Köln
| | - Ulli Umpfenbach
- Arzt für Kinderheilkunde und Jugendmedizin, Kinderpneumologie, Umweltmedizin, klassische Homöopathie, Asthmatrainer, Neurodermitistrainer, Viersen
| | | | | | - Birgit Willinger
- Klinisches Institut für Labormedizin, Klinische Abteilung für Klinische Mikrobiologie - MedUni Wien
| | - Gerhard A Wiesmüller
- Labor Dr. Wisplinghoff
- ZfMK - Zentrum für Umwelt, Hygiene und Mykologie, Köln
- Institut für Arbeits-, Sozial- und Umweltmedizin, Uniklinik RWTH Aachen
| |
Collapse
|
26
|
Zhang M, Wang J, Liu R, Wang Q, Qin S, Chen Y, Li W. The role of Keap1-Nrf2 signaling pathway in the treatment of respiratory diseases and the research progress on targeted drugs. Heliyon 2024; 10:e37326. [PMID: 39309822 PMCID: PMC11414506 DOI: 10.1016/j.heliyon.2024.e37326] [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: 05/28/2024] [Revised: 07/30/2024] [Accepted: 09/01/2024] [Indexed: 09/25/2024] Open
Abstract
Lungs are exposed to external oxidants from the environment as in harmful particles and smog, causing oxidative stress in the lungs and consequently respiratory ailment. The NF-E2-related factor 2 (Nrf2) is the one with transcriptional regulatory function, while its related protein Kelch-like ECH-associated protein 1 (Keap1) inhibits Nrf2 activity. Together, they form the Keap1-Nrf2 pathway, which regulates the body's defense against oxidative stress. This pathway has been shown to maintain cellular homeostasis during oxidative stressing, inflammation, oncogenesis, and apoptosis by coordinating the expression of cytoprotective genes and making it a potential therapeutic target for respiratory diseases. This paper summarizes this point in detail in Chapter 2. In addition, this article summarizes the current drug development and clinical research progress related to the Keap1-Nrf2 signaling pathway, with a focus on the potential of Nrf2 agonists in treating respiratory diseases. Overall, the article reviews the regulatory mechanisms of the Keap1-Nrf2 signaling pathway in respiratory diseases and the progress of targeted drug research, aiming to provide new insights for treatment.
Collapse
Affiliation(s)
- Mengyang Zhang
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, Shandong, 266112, China
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Jing Wang
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Runze Liu
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Qi Wang
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, Shandong, 266112, China
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Song Qin
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, Shandong, 266112, China
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| | - Yuqin Chen
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, 92093, USA
| | - Wenjun Li
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, Shandong, 266112, China
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, Shandong, 264003, China
| |
Collapse
|
27
|
Luo X, Zeng W, Tang J, Liu W, Yang J, Chen H, Jiang L, Zhou X, Huang J, Zhang S, Du L, Shen X, Chi H, Wang H. Multi-modal transcriptomic analysis reveals metabolic dysregulation and immune responses in chronic obstructive pulmonary disease. Sci Rep 2024; 14:22699. [PMID: 39349929 PMCID: PMC11442962 DOI: 10.1038/s41598-024-71773-w] [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: 01/30/2024] [Accepted: 08/30/2024] [Indexed: 10/04/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD), a progressive inflammatory condition of the airways, emerges from the complex interplay between genetic predisposition and environmental factors. Notably, its incidence is on the rise, particularly among the elderly demographic. Current research increasingly highlights cellular senescence as a key driver in chronic lung pathologies. Despite this, the detailed mechanisms linking COPD with senescent genomic alterations remain elusive. To address this gap, there is a pressing need for comprehensive bioinformatics methodologies that can elucidate the molecular intricacies of this link. This approach is crucial for advancing our understanding of COPD and its association with cellular aging processes. Utilizing a spectrum of advanced bioinformatics techniques, this research delved into the potential mechanisms linking COPD with aging-related genes, identifying four key genes (EP300, MTOR, NFE2L1, TXN) through machine learning and weighted gene co-expression network analysis (WGCNA) analyses. Subsequently, a precise diagnostic model leveraging an artificial neural network was developed. The study further employed single-cell analysis and molecular docking to investigate senescence-related cell types in COPD tissues, particularly focusing on the interactions between COPD and NFE2L1, thereby enhancing the understanding of COPD's molecular underpinnings. Leveraging artificial neural networks, we developed a robust classification model centered on four genes-EP300, MTOR, NFE2L1, TXN-exhibiting significant predictive capability for COPD and offering novel avenues for its early diagnosis. Furthermore, employing various single-cell analysis techniques, the study intricately unraveled the characteristics of senescence-related cell types in COPD tissues, enriching our understanding of the disease's cellular landscape. This research anticipates offering novel biomarkers and therapeutic targets for early COPD intervention, potentially alleviating the disease's impact on individuals and healthcare systems, and contributing to a reduction in global COPD-related mortality. These findings carry significant clinical and public health ramifications, bolstering the foundation for future research and clinical strategies in managing and understanding COPD.
Collapse
Affiliation(s)
- Xiufang Luo
- Geriatric Department, Dazhou Central Hospital, Dazhou, 635000, China
| | - Wei Zeng
- Oncology Department, Second People's Hospital of Yaan City, Yaan, 625000, China
| | - Jingyi Tang
- Department of Clinical Medicine, Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Wang Liu
- Department of General Surgery, Cheng Fei Hospital, Chengdu, 610000, China
| | - Jinyan Yang
- School of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Haiqing Chen
- Department of Clinical Medicine, Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Lai Jiang
- Department of Clinical Medicine, Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Xuancheng Zhou
- Department of Clinical Medicine, Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Jinbang Huang
- Department of Clinical Medicine, Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Shengke Zhang
- Department of Clinical Medicine, Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Linjuan Du
- Oncology Department, Dazhou Central Hospital, Dazhou, 635000, China
| | - Xiang Shen
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Hao Chi
- Department of Clinical Medicine, Clinical Medical College, Southwest Medical University, Luzhou, 646000, China.
| | - Huachuan Wang
- Department of Thoracic Surgery, Dazhou Central Hospital, Dazhou, 635000, China.
| |
Collapse
|
28
|
Tian TL, Zhi TY, Xie ML, Jiang YL, Qu XK. Dietary inflammatory index and all-cause mortality in adults with COPD: a prospective cohort study from the NHANES 1999-2018. Front Nutr 2024; 11:1421450. [PMID: 39385783 PMCID: PMC11463153 DOI: 10.3389/fnut.2024.1421450] [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: 04/22/2024] [Accepted: 08/19/2024] [Indexed: 10/12/2024] Open
Abstract
Background Chronic inflammation is closely linked to Chronic Obstructive Pulmonary Disease (COPD); however, the impact of the Dietaryq Inflammatory Index (DII) on mortality among COPD patients remains uncertain. Objective To assess the correlation between the DII and all-cause mortality in COPD patients using data from the National Health and Nutrition Examination Survey (NHANES). Methods We conducted a retrospective cohort study on 1,820 COPD patients from the NHANES dataset (1999-2018). The influence of DII on mortality was evaluated using multivariate Cox regression, smoothing spline fitting, and threshold effect analysis. Additionally, Kaplan-Meier survival analysis was performed to compare survival curves among different DII groups. Subgroup analyses and E-values identified sensitive cohorts and assessed unmeasured confounding. Results Over an average follow-up of 91 months, multivariate Cox regression models revealed a significant positive correlation between DII scores and mortality risk, with each unit increase in DII associated with a 10% higher risk of death (HR: 1.10, 95% CI: 1.03-1.16; P = 0.002). Among the DII tertiles, individuals in the second tertile (T2: 1.23-2.94) experienced a 67% increase in mortality risk compared to those in the lowest tertile (T1: -5.28-1.23) (HR: 1.67, 95% CI: 1.26-2.21; p < 0.001). The third tertile (T3) did not show a statistically significant increase in mortality risk (HR: 1.30, 95% CI: 0.98-1.72; p=0.074). A restricted cubic spline analysis indicated a significant nonlinear association between DII and all-cause mortality (p = 0.021). Threshold effect analysis further revealed that below a DII of 2.19, there was a significant increase in all-cause mortality risk (HR = 1.19, 95% CI: 1.07-1.33; p = 0.002), while at or above this threshold, the risk increase was not statistically significant (HR=0.89, 95% CI: 0.68-1.15; p = 0.380). Kaplan-Meier analysis revealed significant differences in survival curves among DII tertiles (p < 0.001), with the lowest DII tertile showing the highest survival probability. Both subgroup and sensitivity analyses confirmed the robustness of these findings. Conclusion DII is positively correlated with mortality risk in COPD patients, showing nonlinear characteristics and threshold effects, underscoring its prognostic value.
Collapse
Affiliation(s)
- Tu-Lei Tian
- Department of Respiratory and Critical Care Medicine, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, China
| | - Tian-Yu Zhi
- Department of Clinical Medicine (5+3 Integrated Program), Shanxi Medical University, Taiyuan, China
| | - Mei-Ling Xie
- Bengbu Medical University Graduate School, Bengbu, Anhui, China
| | - Ya-Lin Jiang
- Department of Respiratory and Critical Care Medicine, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, China
| | - Xiang-Kun Qu
- Department of Respiratory and Critical Care Medicine, The Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, China
| |
Collapse
|
29
|
de Hilster RHJ, Reinders-Luinge MA, Schuil A, Borghuis T, Harmsen MC, Burgess JK, Hylkema MN. A 3D Epithelial-Mesenchymal Co-Culture Model of the Airway Wall Using Native Lung Extracellular Matrix. Bioengineering (Basel) 2024; 11:946. [PMID: 39329688 PMCID: PMC11428669 DOI: 10.3390/bioengineering11090946] [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: 08/03/2024] [Revised: 09/02/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic lung disease characterized by ongoing inflammation, impaired tissue repair, and aberrant interplay between airway epithelium and fibroblasts, resulting in an altered extracellular matrix (ECM) composition. The ECM is the three-dimensional (3D) scaffold that provides mechanical support and biochemical signals to cells, now recognized not only as a consequence but as a potential driver of disease progression. To elucidate how the ECM influences pathophysiological changes occurring in COPD, in vitro models are needed that incorporate the ECM. ECM hydrogels are a novel experimental tool for incorporating the ECM in experimental setups. We developed an airway wall model by combining lung-derived ECM hydrogels with a co-culture of primary human fibroblasts and epithelial cells at an air-liquid interface. Collagen IV and a mixture of collagen I, fibronectin, and bovine serum albumin were used as basement membrane-mimicking coatings. The model was initially assembled using porcine lung-derived ECM hydrogels and subsequently with COPD and non-COPD human lung-derived ECM hydrogels. The resulting 3D construct exhibited considerable contraction and supported co-culture, resulting in a differentiated epithelial layer. This multi-component 3D model allows the investigation of remodelling mechanisms, exploring ECM involvement in cellular crosstalk, and holds promise as a model for drug discovery studies exploring ECM involvement in cellular interactions.
Collapse
Affiliation(s)
- Roderick H. J. de Hilster
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Marjan A. Reinders-Luinge
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
| | - Annemarie Schuil
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
| | - Theo Borghuis
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Martin C. Harmsen
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
- KOLFF Institute—REGENERATE, University of Groningen, University Medical Center Groningen, FB41, 9713 AV Groningen, The Netherlands
| | - Janette K. Burgess
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- KOLFF Institute—REGENERATE, University of Groningen, University Medical Center Groningen, FB41, 9713 AV Groningen, The Netherlands
| | - Machteld N. Hylkema
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (R.H.J.d.H.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
30
|
Qi Y, Yan Y, Tang D, Han J, Zhu X, Cui M, Wu H, Tao Y, Fan F. Inflammatory and Immune Mechanisms in COPD: Current Status and Therapeutic Prospects. J Inflamm Res 2024; 17:6603-6618. [PMID: 39318994 PMCID: PMC11421452 DOI: 10.2147/jir.s478568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) currently ranks among the top three causes of mortality worldwide, presenting as a prevalent and complex respiratory ailment. Ongoing research has underscored the pivotal role of immune function in the onset and progression of COPD. The immune response in COPD patients exhibits abnormalities, characterized by diminished anti-infection capacity due to immune senescence, heightened activation of neutrophils and macrophages, T cell infiltration, and aberrant B cell activity, collectively contributing to airway inflammation and lung injury in COPD. Objective This review aimed to explore the pivotal role of the immune system in COPD and its therapeutic potential. Methods We conducted a review of immunity and COPD published within the past decade in the Web of Science and PubMed databases, sorting through and summarizing relevant literature. Results This article examines the pivotal roles of the immune system in COPD. Understanding the specific functions and interactions of these immune cells could facilitate the development of novel therapeutic strategies and interventions aimed at controlling inflammation, enhancing immune function, and mitigating the impact of respiratory infections in COPD patients.
Collapse
Affiliation(s)
- Yanan Qi
- School of Pharmacy, Bengbu Medical University, Bengbu, People’s Republic of China
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, People’s Republic of China
| | - Yuanyuan Yan
- School of Pharmacy, Bengbu Medical University, Bengbu, People’s Republic of China
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, People’s Republic of China
| | - Dawei Tang
- School of Pharmacy, Bengbu Medical University, Bengbu, People’s Republic of China
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, People’s Republic of China
| | - Jingjing Han
- School of Pharmacy, Bengbu Medical University, Bengbu, People’s Republic of China
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, People’s Republic of China
| | - Xinyi Zhu
- School of Pharmacy, Bengbu Medical University, Bengbu, People’s Republic of China
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, People’s Republic of China
| | - Mengting Cui
- School of Pharmacy, Bengbu Medical University, Bengbu, People’s Republic of China
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, People’s Republic of China
| | - Hongyan Wu
- Institute of Biomedical Technology, Jiangsu Vocational College of Medicine, Yancheng, Jiangsu, 224005, People’s Republic of China
| | - Yu Tao
- School of Pharmacy, Bengbu Medical University, Bengbu, People’s Republic of China
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, People’s Republic of China
| | - Fangtian Fan
- School of Pharmacy, Bengbu Medical University, Bengbu, People’s Republic of China
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, People’s Republic of China
| |
Collapse
|
31
|
Xu J, Zhu FM, Liu Y, Fang P, Sun J, Liu MY, Tang MM, Zhao H, Fu L, Yang J. Blood cadmium concentration and pulmonary function injury: potential mediating role of oxidative stress in chronic obstructive pulmonary disease patients. BMC Pulm Med 2024; 24:459. [PMID: 39289625 PMCID: PMC11409541 DOI: 10.1186/s12890-024-03269-2] [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: 01/15/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Exposure to cadmium (Cd) is associated with a reduction in lung function among patients with chronic obstructive pulmonary disease (COPD). The longitudinal relationship and mechanism underlying the link between Cd exposure and lung function changes among COPD patients are yet unknown. METHODS The cohort study included 259 eligible patients who underwent regular professional follow-ups. Blood Cd levels and serum 8-iso-prostaglandin F2 alpha (8-iso-PGF2α) levels were assessed. Lung function was determined at baseline and follow-up research. The associations between changes in lung function and blood Cd concentration were analysed using multivariate linear and logistic regression models. RESULTS Each 1-ppb elevation in blood Cd content resulted in a 0.420 L decrease in forced vital capacity (FVC), a 0.424 L decrease in forced expiratory volume in 1 s (FEV1), a 4.341% decrease in FEV1/FVC%, and a 8.418% decrease in FEV1% predicted in patients with COPD. Blood Cd concentration showed a positive correlation with serum 8-iso-PGF2α levels in a specific range. The relative contribution of increased serum levels of 8-iso-PGF2α to Cd-induced declines in FEV1, predicted FEV1%, and FEV1/FVC% were 2.08%, 8.08%, and 13.19%, respectively. CONCLUSION Blood Cd levels are associated with lung function changes in COPD patients. Oxidative stress is thought to be an important mediator in Cd-induced reduction of pulmonary function.
Collapse
Affiliation(s)
- Juan Xu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
- Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Feng-Min Zhu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
- Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Ying Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
- Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Pu Fang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
- Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Jing Sun
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
- Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Ming-Yan Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
- Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Min-Min Tang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
- Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Hui Zhao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
- Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
- Centre for Big Data and Population Health of IHM, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Lin Fu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China.
- Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China.
- Centre for Big Data and Population Health of IHM, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China.
| | - Jin Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China.
- Institute of Respiratory Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China.
- Centre for Big Data and Population Health of IHM, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China.
- Information Management Centre, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China.
| |
Collapse
|
32
|
Wang J, Wang R, Li Y, Huang J, Liu Y, Wang J, Xian P, Zhang Y, Yang Y, Zhang H, Li J. Lipolysis engages CD36 to promote ZBP1-mediated necroptosis-impairing lung regeneration in COPD. Cell Rep Med 2024; 5:101732. [PMID: 39255796 PMCID: PMC11525022 DOI: 10.1016/j.xcrm.2024.101732] [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: 12/07/2023] [Revised: 06/05/2024] [Accepted: 08/19/2024] [Indexed: 09/12/2024]
Abstract
Lung parenchyma destruction represents a severe condition commonly found in chronic obstructive pulmonary disease (COPD), a leading cause of morbidity and mortality worldwide. Promoting lung regeneration is crucial for achieving clinical improvement. However, no therapeutic drugs are approved to improve the regeneration capacity due to incomplete understanding of the underlying pathogenic mechanisms. Here, we identify a positive feedback loop formed between adipose triglyceride lipase (ATGL)-mediated lipolysis and overexpression of CD36 specific to lung epithelial cells, contributing to disease progression. Genetic deletion of CD36 in lung epithelial cells and pharmacological inhibition of either ATGL or CD36 effectively reduce COPD pathogenesis and promote lung regeneration in mice. Mechanistically, disruption of the ATGL-CD36 loop rescues Z-DNA binding protein 1 (ZBP1)-induced cell necroptosis and restores WNT/β-catenin signaling. Thus, we uncover a crosstalk between lipolysis and lung epithelial cells, suggesting the regenerative potential for therapeutic intervention by targeting the ATGL-CD36-ZBP1 axis in COPD.
Collapse
Affiliation(s)
- Jiazhen Wang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Ru Wang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yicun Li
- Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jiahui Huang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yang Liu
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jiayi Wang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Peng Xian
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yuanhang Zhang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yanmei Yang
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Haojian Zhang
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China; Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| | - Jiansheng Li
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, China; Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, China; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China.
| |
Collapse
|
33
|
Feng Y, Zhang W, Bao S, Shen J. Active Components of Wen Fei Fu Yang Qu Tan Fang and its Molecular Targets for Chronic Obstructive Pulmonary Disease Based on Network Pharmacology and Molecular Docking. Cell Biochem Biophys 2024:10.1007/s12013-024-01498-0. [PMID: 39259410 DOI: 10.1007/s12013-024-01498-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2024] [Indexed: 09/13/2024]
Abstract
To investigate the mechanism of Wen Fei Fu Yang Qu Tan Fang (WFFYQTF) in the treatment of chronic obstructive pulmonary disease (COPD) using network pharmacology and pharmacodynamics. The TCMSP database was utilized to identify the chemical components and molecular targets of WFFYQTF. Cytoscape software was employed to construct a "drug component-target" network. COPD risk genes and intersecting molecular targets of WFFYQTF were identified using GeneCards, OMIM, and DisGeNET databases. The STRING website was the place where protein-protein interaction (PPI) analysis was performed. Cytoscape topological analysis was applied for screening out key targets of WFFYQTF. GO and KEGG enrichment analyses were conducted using the DAVID database to elucidate the treatment targets of COPD with WFFYQTF. A total of 136 active components of WFFYQTF were identified, including key components such as quercetin, kaempferol, and luteolin, which were found to be particularly significant. Additionally, 412 drug targets and 7121 COPD risk genes were screened out, and 323 treatment targets of COPD with WFFYQTF were determined by Wayne analysis. Core targets identified via PPI analysis included SRC, STAT3, AKT1, HSP90AA1, and JUN. Pathways such as the hypoxia responce, inflammatory response, PI3K/AKT pathway, TH17 pathway and MAPK pathway were obtained with GO and KEGG enrichment analyses. Molecular docking results suggested that quercetin could be soundly bound to STAT3 and AKT1, and kaempferol to SRC. WFFYQTF can effectively impede COPD progression through the coordinated action of multiple components, targets, and pathways during treatment.
Collapse
Affiliation(s)
- Yangrong Feng
- Department of Classical Internal Medicine of TCM, Zhejiang Chinese Medicine University, Ningbo, Zhejiang Province, China
| | - Wei Zhang
- Department of Emergency Medicine, Ningbo Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medicine University, Ningbo, Zhejiang Province, China
| | - Sanyu Bao
- Department of Classical Internal Medicine of TCM, Zhejiang Chinese Medicine University, Ningbo, Zhejiang Province, China
| | - Jieru Shen
- Department of Classical Internal Medicine of TCM, Zhejiang Chinese Medicine University, Ningbo, Zhejiang Province, China.
| |
Collapse
|
34
|
Wen Z, Ablimit A. Comprehensive analysis of scRNA-Seq and bulk RNA-Seq reveals ubiquitin promotes pulmonary fibrosis in chronic pulmonary diseases. Sci Rep 2024; 14:21195. [PMID: 39261509 PMCID: PMC11390722 DOI: 10.1038/s41598-024-70659-1] [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: 11/16/2023] [Accepted: 08/20/2024] [Indexed: 09/13/2024] Open
Abstract
It is estimated that there are 544.9 million people suffering from chronic respiratory diseases in the world, which is the third largest chronic disease. Although there are various clinical treatment methods, there is no specific drug for chronic pulmonary diseases, including chronic obstructive pulmonary disease (COPD), interstitial lung disease (ILD) and idiopathic pulmonary fibrosis (IPF). Therefore, it is urgent to clarify the pathological mechanism and medication development. Single-cell transcriptome data of human and mouse from GEO database were integrated by "Harmony" algorithm. The data was standardized and normalized by using "Seurat" package, and "SingleR" algorithm was used for cell grouping annotation. The "Findmarker" function is used to find differentially expressed genes (DEGs), which were enriched and analyzed by using "clusterProfiler", and a protein interaction network was constructed for DEGs, and four algorithms are used to find the hub genes. The expression of hub genes were analyzed in independent human and mouse single-cell transcriptome data. Bulk RNA data were used to integrate by the "SVA" function, verify the expression levels of hub genes and build a diagnostic model. The L1000FWD platform was used to screen potential drugs. Through exploring the similarities and differences by integrated single-cell atlas, we found that the lung parenchymal cells showed abnormal oxidative stress, cell matrix adhesion and ubiquitination in COPD, corona virus disease 2019 (COVID-19), ILD and IPF. Meanwhile, the lung resident immune cells showed abnormal Toll-like receptor signals, interferon signals and ubiquitination. However, unlike acute pneumonia (COVID-19), chronic pulmonary disease shows enhanced ubiquitination. This phenomenon was confirmed in independent external human single-cell atlas, but unfortunately, it was not confirmed in mouse single-cell atlas of bleomycin-induced pulmonary fibrosis model and influenza virus-infected mouse model, which means that the model needs to be optimized. In addition, the bulk RNA-Seq data of COVID-19, ILD and IPF was integrated, and we found that the immune infiltration of lung tissue was enhanced, consistent with the single-cell level, UBA52, UBB and UBC were low expressed in COVID-19 and high expressed in ILD, and had a strong correlation with the expression of cell matrix adhesion genes. UBA52 and UBB have good diagnostic efficacy, and salermide and SSR-69071 can be used as their candidate drugs. Our study found that the disorder of protein ubiquitination in chronic pulmonary diseases is an important cause of pathological phenotype of pulmonary fibrosis by integrating scRNA-Seq and bulk RNA-Seq, which provides a new horizons for clinicopathology, diagnosis and treatment.
Collapse
Affiliation(s)
- Zhuman Wen
- Department of Histology and Embryology, Basic Medical College, Xinjiang Medical University, Ürümqi, China
- College of Nursing and Health, Xinjiang Career Technical College, Kuitun, China
| | - Abduxukur Ablimit
- Department of Histology and Embryology, Basic Medical College, Xinjiang Medical University, Ürümqi, China.
| |
Collapse
|
35
|
Wu R, Gong H. The association between non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio and chronic obstructive pulmonary disease: the mediating role of dietary inflammatory index. Front Nutr 2024; 11:1427586. [PMID: 39315013 PMCID: PMC11416962 DOI: 10.3389/fnut.2024.1427586] [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: 05/04/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
Background Numerous studies have indicated a potential correlation between COPD, lipid metabolism, and dietary inflammation. However, the exact mechanisms by which dietary inflammation regulates the pathological processes of COPD related to lipid metabolism remain unclear. NHHR is a novel composite index of atherosclerotic lipid profiles, while the Dietary Inflammatory Index (DII) measures diet-induced inflammation. This study explores the relationship between NHHR and COPD and evaluates whether DII mediates this association. Methods We employed multivariable logistic regression, smooth curve fitting, threshold effect analysis, and subgroup analysis to explore the relationship between NHHR and the incidence of COPD. Additionally, we conducted a mediation analysis to explore the potential relationship between dietary inflammatory index (DII) levels and the relationship between NHHR and COPD. Results This analysis encompassed 13,452 participants, with 2,332 reporting incidents of COPD. Following adjustment for all covariates using multivariable logistic regression, each unit increase in NHHR level and DII level was associated with a 10% (OR = 1.10, 95% CI: 1.05, 1.16) and 8% (OR = 1.08, 95% CI: 1.04, 1.13) increase, respectively, in the incidence rate of COPD. Furthermore, compared to the lowest quartile, the highest quartile of NHHR level and DII level was associated with a 47% (p < 0.001) and 50% (p < 0.001) increase, respectively, in the incidence rate of COPD. Smooth curve fitting and threshold effect analysis revealed a nonlinear relationship between NHHR and the risk of COPD, with a breakpoint at 2.60. Mediation analysis indicated that DII mediated 7.24% of the association between NHHR and COPD (p = 0.004). Conclusion Higher NHHR levels are associated with an increased prevalence of COPD. Moreover, this association is mediated by DII, suggesting that an anti-inflammatory diet may be beneficial.
Collapse
Affiliation(s)
- Ruying Wu
- Department of Surgery 3, Hebei Provincial First Veterans Hospital (Hebei General Hospital for Veterans), Xingtai, Hebei, China
| | - Hongyang Gong
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Republic of Korea
| |
Collapse
|
36
|
Huang Y, Hu B, Chen S, Jiang Z, Dai Z, Jiang Z, Liu C, Xu Y, Chen X, Jin W, Yu B, Zhang X. The role of serum chloride ion in the prognosis of COPD. Am J Med Sci 2024; 368:235-241. [PMID: 38777153 DOI: 10.1016/j.amjms.2024.05.006] [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: 04/21/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND As exacerbations of chronic obstructive pulmonary disease (COPD) are one of the leading causes of hospitalization and are associated with significant mortality, it is particularly important to accurately assess the risk of exacerbations in COPD. Most of the current clinical biomarkers are related to inflammation and few consider how ion levels affect COPD. Chloride ion, the second most abundant serum electrolyte, has been shown to be associated with poor prognoses in several diseases, but their relationship with COPD remains unclear. METHODS In total, 105 patients with acute exacerbations of COPD were recruited. Data on clinical characteristics, lung function, blood count, blood biochemistry, relevant scales including the Clinical COPD Questionnaire (CCQ), BODE (BMI, airflow obstruction, dyspnea, exercise capacity) index and the St. George's Respiratory Questionnaire (SGRQ) were collected from all patients for statistical analysis. RESULT There were significant differences in lung function indicators and disease severity in the low chloride ion subgroup compared with the high chloride ion subgroup. On multiple logistic regression analysis, chloride ion was an independent factor affecting lung function in COPD patients (OR=0.808, 95% CI: 0.708 - 0.922, p=0.002). The sensitivity of chloride ion in predicting COPD severity was 78%, the specificity was 63%, and the area under the curve was 0.734 (p<0.001). Subgroup analysis showed that chloride ion was a stronger predictor in male and smoking patients. CONCLUSIONS Chloride ion was a novel prognostic biomarker for COPD, and low levels of chloride ion were independently associated with exacerbations in COPD patients.
Collapse
Affiliation(s)
- Yiben Huang
- Department of Respiratory and Critical Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Binbin Hu
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, China
| | - Siyao Chen
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, China
| | - Zerui Jiang
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, China
| | - Zicong Dai
- The First School of Medicine, School of Information and Engineering, Wenzhou Medical University, Wenzhou, China
| | - Zihan Jiang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chunyan Liu
- Department of Respiratory and Critical Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yage Xu
- Department of Respiratory and Critical Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xianjing Chen
- Department of Respiratory and Critical Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wanzhong Jin
- Department of Respiratory and Critical Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Beibei Yu
- Department of Respiratory and Critical Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaodiao Zhang
- Department of Respiratory and Critical Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
37
|
Kim YC, Sohn KH, Kang HR. Gut microbiota dysbiosis and its impact on asthma and other lung diseases: potential therapeutic approaches. Korean J Intern Med 2024; 39:746-758. [PMID: 39252487 PMCID: PMC11384250 DOI: 10.3904/kjim.2023.451] [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: 10/21/2023] [Revised: 03/08/2024] [Accepted: 04/07/2024] [Indexed: 09/11/2024] Open
Abstract
The emerging field of gut-lung axis research has revealed a complex interplay between the gut microbiota and respiratory health, particularly in asthma. This review comprehensively explored the intricate relationship between these two systems, focusing on their influence on immune responses, inflammation, and the pathogenesis of respiratory diseases. Recent studies have demonstrated that gut microbiota dysbiosis can contribute to asthma onset and exacerbation, prompting investigations into therapeutic strategies to correct this imbalance. Probiotics and prebiotics, known for their ability to modulate gut microbial compositions, were discussed as potential interventions to restore immune homeostasis. The impact of antibiotics and metabolites, including short-chain fatty acids produced by the gut microbiota, on immune regulation was examined. Fecal microbiota transplantation has shown promise in various diseases, but its role in respiratory disorders is not established. Innovative approaches, including mucus transplants, inhaled probiotics, and microencapsulation strategies, have been proposed as novel therapeutic avenues. Despite challenges, including the sophisticated adaptability of microbial communities and the need for mechanistic clarity, the potential for microbiota-based interventions is considerable. Collaboration between researchers, clinicians, and other experts is essential to unravel the complexities of the gut-lung axis, paving a way for innovative strategies that could transform the management of respiratory diseases.
Collapse
Affiliation(s)
- Young-Chan Kim
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kyoung-Hee Sohn
- Division of Respiratory, Allergy and Critical Care Medicine, Department of Internal Medicine, Kyung Hee University Hospital, Seoul, Korea
| | - Hye-Ryun Kang
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
38
|
Tang F, Liu HY, He QY, Liu Y, Lv LP, Fei J, Fu L. Cobalt exposure and pulmonary function reduction in chronic obstructive pulmonary disease patients: the mediating role of club cell secretory protein. Respir Res 2024; 25:324. [PMID: 39182083 PMCID: PMC11344942 DOI: 10.1186/s12931-024-02950-8] [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: 03/17/2024] [Accepted: 08/11/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Cobalt (Co) is a metal which is widely used in the industrial production. The previous studies found the toxic effects of environmental Co exposure on multiple organs. However, the correlation of blood Co concentration with lung function was inconsistent in patients with chronic obstructive pulmonary disease (COPD). METHODS All 771 stable COPD patients were recruited. Peripheral blood and clinical information were collected. The levels of blood Co and serum CC16 were measured. RESULTS Cross-sectional study suggested that the level of blood Co was inversely and dose-dependently related to lung function parameters. Each 1 ppm elevation of blood Co was related to 0.598 L decline in FVC, 0.465 L decline in FEV1, 6.540% decline in FEV1/FVC%, and 14.013% decline in FEV1%, respectively. Moreover, higher age, enrolled in winter, current-smoking, higher smoking amount, and inhaled corticosteroids prominently exacerbated the negative correlation between blood Co and lung function. Besides, serum CC16 content was gradually reduced with blood Co elevation in COPD patients. Besides, serum CC16 was positively correlated with lung function, and inversely related to blood Co. Additionally, decreased CC16 substantially mediated 11.45% and 6.37% Co-triggered downregulations in FEV1 and FEV1%, respectively. CONCLUSION Blood Co elevation is closely related to the reductions of pulmonary function and serum CC16. CC16 exerts a significantly mediating role of Co-related to pulmonary function decrease among COPD patients.
Collapse
Affiliation(s)
- Fei Tang
- Department of Interventional Pulmonology and Endoscopic Diagnosis and Treatment Center, Anhui Chest Hospital, Hefei, 230022, Anhui, China
| | - Hong-Yan Liu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Furong Road no 678, Hefei, 230601, Anhui, China
- Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Qi-Yuan He
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Furong Road no 678, Hefei, 230601, Anhui, China
| | - Ying Liu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Furong Road no 678, Hefei, 230601, Anhui, China
| | - Li-Ping Lv
- Department of Interventional Pulmonology and Endoscopic Diagnosis and Treatment Center, Anhui Chest Hospital, Hefei, 230022, Anhui, China.
| | - Jun Fei
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Furong Road no 678, Hefei, 230601, Anhui, China.
- Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
| | - Lin Fu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Furong Road no 678, Hefei, 230601, Anhui, China.
- Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China.
- Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, 230032, Anhui, China.
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Bengbu Medical College, Bengbu, 233030, Anhui, China.
| |
Collapse
|
39
|
Sweef O, Mahfouz R, Taşcıoğlu T, Albowaidey A, Abdelmonem M, Asfar M, Zaabout E, Corcino YL, Thomas V, Choi ES, Furuta S. Decoding LncRNA in COPD: Unveiling Prognostic and Diagnostic Power and Their Driving Role in Lung Cancer Progression. Int J Mol Sci 2024; 25:9001. [PMID: 39201688 PMCID: PMC11354875 DOI: 10.3390/ijms25169001] [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: 07/01/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer represent formidable challenges in global health, characterized by intricate pathophysiological mechanisms and multifaceted disease progression. This comprehensive review integrates insights from diverse perspectives to elucidate the intricate roles of long non-coding RNAs (lncRNAs) in the pathogenesis of COPD and lung cancer, focusing on their diagnostic, prognostic, and therapeutic implications. In the context of COPD, dysregulated lncRNAs, such as NEAT1, TUG1, MALAT1, HOTAIR, and GAS5, emerge as pivotal regulators of genes involved in the disease pathogenesis and progression. Their identification, profiling, and correlation with the disease severity present promising avenues for prognostic and diagnostic applications, thereby shaping personalized disease interventions. These lncRNAs are also implicated in lung cancer, underscoring their multifaceted roles and therapeutic potential across both diseases. In the domain of lung cancer, lncRNAs play intricate modulatory roles in disease progression, offering avenues for innovative therapeutic approaches and prognostic indicators. LncRNA-mediated immune responses have been shown to drive lung cancer progression by modulating the tumor microenvironment, influencing immune cell infiltration, and altering cytokine production. Their dysregulation significantly contributes to tumor growth, metastasis, and chemo-resistance, thereby emphasizing their significance as therapeutic targets and prognostic markers. This review summarizes the transformative potential of lncRNA-based diagnostics and therapeutics for COPD and lung cancer, offering valuable insights into future research directions for clinical translation and therapeutic development.
Collapse
Affiliation(s)
- Osama Sweef
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
- Department of Zoology, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Reda Mahfouz
- Core Laboratory, University Hospital Cleveland Medical Center, Department of Pathology, School of Medicine, Case Western Reserve University, 1100 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Clinical Pathology, Faculty of Medicine, Menofia University, Shebin-Elkom 32511, Egypt
| | - Tülin Taşcıoğlu
- Department of Molecular Biology and Genetics, Demiroglu Bilim University, Esentepe Central Campus, Besiktas, 34394 Istanbul, Turkey
| | - Ali Albowaidey
- The Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Mohamed Abdelmonem
- Department of Pathology, Transfusion Medicine Service, Stanford Healthcare, Stanford, CA 94305, USA
| | - Malek Asfar
- Department of Pathology, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
| | - Elsayed Zaabout
- Department of Therapeutics & Pharmacology, The University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX 77030, USA
| | - Yalitza Lopez Corcino
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
| | - Venetia Thomas
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
| | - Eun-Seok Choi
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
| | - Saori Furuta
- Division of Cancer Biology, Department of Medicine, MetroHealth Medical Center, School of Medicine, Case Western Reserve University, 2500 MetroHealth Drive, Cleveland, OH 44109, USA
| |
Collapse
|
40
|
Liu P, Li Y, Tang D, Liu G, Zou Y, Ma Y, Zheng W. Effects of different traditional Chinese exercises on pulmonary function in patients with stable chronic obstructive pulmonary disease: a network meta-analysis. BMC Complement Med Ther 2024; 24:304. [PMID: 39143580 PMCID: PMC11325568 DOI: 10.1186/s12906-024-04609-9] [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: 01/30/2024] [Accepted: 08/08/2024] [Indexed: 08/16/2024] Open
Abstract
OBJECTIVE Traditional Chinese exercises (Taichi, Wuqinxi, Liuzijue, and Baduanjin) are considered effective alternative treatments for improving symptoms in the stable phase of COPD. However, the most effective exercise remains unknown. This study compared the effectiveness of different traditional Chinese exercises on pulmonary function in patients with stable chronic obstructive pulmonary disease (COPD) using a network meta-analysis. METHODS From database establishment until September 2023, eligible randomized controlled trials (RCTs) were searched. Two reviewers performed the risk of bias assessment of the included studies using the Cochrane Collaboration tool, and the evidence level was suggested using the GRADE system. RESULTS Fifty-seven studies comprising 4294 patients were included. The results of the network meta-analysis show that Baduanjin was most effective in improving the forced expiratory volume in the first second (FEV1). However, Liuzijue significantly improved the first-second forced vital capacity percentage of expected value (FEV1%) and the ratio of the forced expiratory volume in the first second to the forced vital capacity (FEV1/FVC). The probability ranking results indicated that Liuzijue was the most effective, followed by Baduanjin, Wuqinxi, and Taichi. Subgroup analysis in conjunction with intervention duration revealed that Liuzijue had a significant advantage over other interventions for improving FEV1, FEV1%, and FEV1/FVC within 6 months and improved FEV1% and FEV1/FVC for ≥ 6 months. Moreover, Subgroup analysis based on baseline pulmonary function revealed that Liuzijue had a significant advantage over other interventions for improving FEV1% within severe and moderate groups. Finally, Subgroup analysis based on the frequency of interventions showed that Liuzijue was still more effective in improving FEV1, FEV1%, and FEV1/FVC in the ≥ three times one week. CONCLUSION Liuzijue was more effective than Taichi, Wuqinxi, Liuzijue, and Baduanjin in improving pulmonary function in patients with stable COPD.
Collapse
Affiliation(s)
- Peirong Liu
- Key Laboratory of Sports Engineering of General Administration of Sports of China, Research Center of Sports Equipment Engineering Technology of Hubei Province, Wuhan Sports University, Wuhan, 430079, People's Republic of China
| | - Yongjie Li
- Department of Rehabilitation Medicine, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, 550014, People's Republic of China
| | - Ding Tang
- Key Laboratory of Sports Engineering of General Administration of Sports of China, Research Center of Sports Equipment Engineering Technology of Hubei Province, Wuhan Sports University, Wuhan, 430079, People's Republic of China
| | - Gan Liu
- Key Laboratory of Sports Engineering of General Administration of Sports of China, Research Center of Sports Equipment Engineering Technology of Hubei Province, Wuhan Sports University, Wuhan, 430079, People's Republic of China
| | - Yao Zou
- Key Laboratory of Sports Engineering of General Administration of Sports of China, Research Center of Sports Equipment Engineering Technology of Hubei Province, Wuhan Sports University, Wuhan, 430079, People's Republic of China
| | - Yong Ma
- Key Laboratory of Sports Engineering of General Administration of Sports of China, Research Center of Sports Equipment Engineering Technology of Hubei Province, Wuhan Sports University, Wuhan, 430079, People's Republic of China
| | - Weitao Zheng
- Key Laboratory of Sports Engineering of General Administration of Sports of China, Research Center of Sports Equipment Engineering Technology of Hubei Province, Wuhan Sports University, Wuhan, 430079, People's Republic of China.
| |
Collapse
|
41
|
Kim GD, Shin DU, Song HJ, Lim KM, Eom JE, Lim EY, Kim YI, Song JH, Kim HJ, Lee SY, Shin HS. Analysis of particulate matter-induced alteration of genes and related signaling pathways in the respiratory system. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 281:116637. [PMID: 38941663 DOI: 10.1016/j.ecoenv.2024.116637] [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: 03/28/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024]
Abstract
Airborne particulate matter (PM) is a global environmental risk factor threatening human health and is a major cause of cardiovascular and respiratory disease-associated death. Current studies on PM exposure have been limited to large-scale cohort and epidemiological investigations, emphasizing the need for detailed individual-level studies to uncover specific differentially expressed genes and their associated signaling mechanisms. Herein, we revealed that PM exposure significantly upregulated inflammatory and immune responses, such as cytokine-mediated signaling pathways, complement system, and the activation and migration of immune cells in gene set enrichment analysis of our RNA sequencing (RNAseq) data. Remarkably, we discovered that the broad gene expression and signaling pathways mediated by macrophages were predominantly expressed in the respiratory system following PM exposure. Consistent with these observations, individual PMs, classified by aerodynamic size and origin, significantly promoted macrophage recruitment to the lungs in the mouse lung inflammation model. Additionally, we confirmed that RNAseq observations from the respiratory system were reproduced in murine bone marrow-derived macrophages and the alveolar macrophage cell line MH-S after individual PM exposure. Our findings demonstrated that PM exposure augmented broad inflammatory and immune responses in the respiratory system and suggested the reinforcement of global strategies for reducing particulate air pollution to prevent respiratory diseases and their exacerbation.
Collapse
Affiliation(s)
- Gun-Dong Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Dong-Uk Shin
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Hyeon-Ji Song
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Kyung Min Lim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Ji-Eun Eom
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Eun Yeong Lim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Young In Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Ju Hye Song
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Ha-Jung Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - So-Young Lee
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Hee Soon Shin
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea; Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea.
| |
Collapse
|
42
|
Hua Y, Fan X, Yang M, Su J, Guo J, Jin J, Sun D, Pei P, Yu C, Lyu J, Tao R, Zhou J, Lu Y. Association between socioeconomic status and risk of chronic obstructive pulmonary disease in China: a prospective cohort study. BMC Public Health 2024; 24:2077. [PMID: 39085848 PMCID: PMC11292937 DOI: 10.1186/s12889-024-19490-x] [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: 03/14/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024] Open
Abstract
OBJECTIVE Socioeconomic status (SES) has been proven to be associated with chronic obstructive pulmonary disease (COPD) in Western populations, but the evidence is very limited in China. This study aimed to investigate the association between SES and the risk of COPD incident. METHODS This study was based on the China Kadoorie Biobank (CKB) project in Wuzhong District, Suzhou. A total of 45,484 adults aged 30-79 were included in the analysis during 2004-2008. We used Cox proportional hazard models to investigate the association between SES and the risk of COPD. Household income, education, private property and consumption potential was used to measure SES. Incident COPD cases were ascertained using hospitalization records, death certificates, and active follow-up. RESULTS A total of 524 COPD cases were identified during a median follow-up of 11.2 years. Household income was inversely associated with the risk of COPD (Ptrend<0.005). The adjusted hazard ratios (95% confidence intervals) for incident COPD were 0.88 (0.69-1.14), 0.77 (0.60-0.99), and 0.42 (0.31-0.57) for participants with annual household income of 10,000 ~ 19,999 yuan, 20,000 ~ 34,999 yuan and ≥ 35,000 yuan respectively, in comparison to participants with an annual household income < 10,000 yuan. Furthermore, we found that education level, refrigerator use, private toilet, private phone, and motor vehicle were adversely associated with COPD risk, while ownership of newly renovated flats was positively correlated with COPD incident. CONCLUSIONS This prospective study suggests that SES is associated with the risk of COPD in Chinese adults. Population-based COPD prevention strategies tailored for people with different SES could help reduce the burden of COPD in Chinese.
Collapse
Affiliation(s)
- Yujie Hua
- Department of Non-communicable Chronic Disease Control and Prevention, Suzhou Center for Disease Control and Prevention, Suzhou, 215004, China
| | - Xikang Fan
- Jiangsu Provincial Center for Disease Control and Prevention, 172 Jiangsu Road, Nanjing, 210009, Jiangsu, China
| | - Mengshi Yang
- Department of Epidemiology and Health Statistics, Southeast University, Nanjing, 210009, China
| | - Jian Su
- Jiangsu Provincial Center for Disease Control and Prevention, 172 Jiangsu Road, Nanjing, 210009, Jiangsu, China
| | - Jia Guo
- Department of Non-communicable Chronic Disease Control and Prevention, Suzhou Center for Disease Control and Prevention, Suzhou, 215004, China
| | - Jianrong Jin
- Wuzhong District Disease Control and Prevention Centre, Suzhou, 215000, China
| | - Dianjianyi Sun
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Xueyuan Road, Haidian District, Beijing, 100191, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, 100191, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, 100191, China
| | - Pei Pei
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, 100191, China
| | - Canqing Yu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Xueyuan Road, Haidian District, Beijing, 100191, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, 100191, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, 100191, China
| | - Jun Lyu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Xueyuan Road, Haidian District, Beijing, 100191, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, 100191, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, 100191, China
| | - Ran Tao
- Jiangsu Provincial Center for Disease Control and Prevention, 172 Jiangsu Road, Nanjing, 210009, Jiangsu, China
| | - Jinyi Zhou
- Jiangsu Provincial Center for Disease Control and Prevention, 172 Jiangsu Road, Nanjing, 210009, Jiangsu, China.
| | - Yan Lu
- Department of Non-communicable Chronic Disease Control and Prevention, Suzhou Center for Disease Control and Prevention, Suzhou, 215004, China.
| |
Collapse
|
43
|
Ancer-Rodríguez J, Gopar-Cuevas Y, García-Aguilar K, Chávez-Briones MDL, Miranda-Maldonado I, Ancer-Arellano A, Ortega-Martínez M, Jaramillo-Rangel G. Cell Proliferation and Apoptosis-Key Players in the Lung Aging Process. Int J Mol Sci 2024; 25:7867. [PMID: 39063108 PMCID: PMC11276691 DOI: 10.3390/ijms25147867] [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: 06/18/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Currently, the global lifespan has increased, resulting in a higher proportion of the population over 65 years. Changes that occur in the lung during aging increase the risk of developing acute and chronic lung diseases, such as acute respiratory distress syndrome, chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, and lung cancer. During normal tissue homeostasis, cell proliferation and apoptosis create a dynamic balance that constitutes the physiological cell turnover. In basal conditions, the lungs have a low rate of cell turnover compared to other organs. During aging, changes in the rate of cell turnover in the lung are observed. In this work, we review the literature that evaluates the role of molecules involved in cell proliferation and apoptosis in lung aging and in the development of age-related lung diseases. The list of molecules that regulate cell proliferation, apoptosis, or both processes in lung aging includes TNC, FOXM1, DNA-PKcs, MicroRNAs, BCL-W, BCL-XL, TCF21, p16, NOX4, NRF2, MDM4, RPIA, DHEA, and MMP28. However, despite the studies carried out to date, the complete signaling pathways that regulate cell turnover in lung aging are still unknown. More research is needed to understand the changes that lead to the development of age-related lung diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Gilberto Jaramillo-Rangel
- Department of Pathology, School of Medicine, Autonomous University of Nuevo León, Monterrey 64460, Mexico; (J.A.-R.); (Y.G.-C.); (M.-d.-L.C.-B.); (I.M.-M.); (A.A.-A.); (M.O.-M.)
| |
Collapse
|
44
|
Bae J, Lee HJ, Choi KY, Lee JK, Park TY, Heo EY, Lee CH, Kim DK, Lee HW. Risk factors of acute exacerbation and disease progression in young patients with COPD. BMJ Open Respir Res 2024; 11:e001740. [PMID: 39019624 PMCID: PMC11256056 DOI: 10.1136/bmjresp-2023-001740] [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: 04/01/2023] [Accepted: 06/28/2024] [Indexed: 07/19/2024] Open
Abstract
OBJECTIVE We aimed to elucidate the clinical factors associated with acute exacerbation and disease progression in young patients with chronic obstructive pulmonary disease (COPD). METHODS This retrospective longitudinal observational study included patients with COPD aged between 20 and 50 years with post-bronchodilator forced expiratory volume in one second (FEV1)/forced vital capacity (FVC)<0.7. Eligible patients were followed up with ≥2 spirometry examinations at 1 year interval after COPD diagnosis. The primary outcome was moderate-to-severe acute exacerbation in young patients with COPD. Secondary outcomes were early initiation of regular inhalation therapy and accelerated annual post-bronchodilator FEV1 decline. RESULTS A total of 342 patients were followed up during a median of 64 months. In multivariable analyses, risk factors for moderate-to-severe exacerbation were history of asthma (adjusted HR (aHR)=2.999, 95% CI=[2.074-4.335]), emphysema (aHR=1.951, 95% CI=[1.331-2.960]), blood eosinophil count >300/µL (aHR=1.469, 95% CI=[1.038-2.081]) and low FEV1 (%) (aHR=0.979, 95% CI=[0.970-0.987]). A history of asthma, sputum, blood eosinophil count >300/µL, low FEV1 (%) and low diffusing capacity of the lung for carbon monoxide (DLCO) (%) were identified as clinical factors associated with the early initiation of regular inhalation therapy. The risk factors associated with worsened FEV1 decline were increasing age, female sex, history of pulmonary tuberculosis, sputum, low FEV1 (%) and low DLCO (%). CONCLUSIONS In young COPD patients, specific high-risk features of acute exacerbation and disease progression need to be identified, including a history of previous respiratory diseases, current respiratory symptoms, blood eosinophil counts, and structural or functional pulmonary impairment.
Collapse
Affiliation(s)
- Juye Bae
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Jongno-gu, Seoul, Korea (the Republic of)
| | - Hyo Jin Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Dongjak-gu, Seoul, Korea (the Republic of)
| | - Kwang Yong Choi
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang-si, Korea (the Republic of)
| | - Jung-Kyu Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Dongjak-gu, Seoul, Korea (the Republic of)
| | - Tae Yun Park
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Dongjak-gu, Seoul, Korea (the Republic of)
| | - Eun Young Heo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Dongjak-gu, Seoul, Korea (the Republic of)
| | - Chang Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Hospital, Jongno-gu, Seoul, Korea (the Republic of)
| | - Deog Kyeom Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Dongjak-gu, Seoul, Korea (the Republic of)
| | - Hyun Woo Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Dongjak-gu, Seoul, Korea (the Republic of)
| |
Collapse
|
45
|
Wang M, Zhu M, Jia X, Wu J, Yuan Q, Xu T, Wang Z, Huang M, Ji N, Zhang M. LincR-PPP2R5C regulates IL-1β ubiquitination in macrophages and promotes airway inflammation and emphysema in a murine model of COPD. Int Immunopharmacol 2024; 139:112680. [PMID: 39018689 DOI: 10.1016/j.intimp.2024.112680] [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: 11/17/2023] [Revised: 06/15/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a common disease with high global morbidity and mortality. Macrophages release IL-1β and orchestrate airway inflammation in COPD. Previously, we explored the role of a new lncRNA, LincR-PPP2R5C, in regulating Th2 cells in asthma. Here, we established a murine model of COPD and explored the roles and mechanisms by which LincR-PPP2R5C regulates IL-1β in macrophages. LincR-PPP2R5C was highly expressed in pulmonary macrophages from COPD-like mice. LincR-PPP2R5C deficiency ameliorated emphysema and pulmonary inflammation, as characterized by reduced IL-1β in macrophages. Unexpectedly, in both lung tissues and macrophages, LincR-PPP2R5C deficiency decreased the expression of the IL-1β protein but not the IL-1β mRNA. Furthermore, we found that LincR-PPP2R5C deficiency increased the level of ubiquitinated IL-1β in macrophages, which was mediated by PP2A activity. Targeting PP2A with FTY720 decreased IL-1β and improved COPD. In conclusion, LincR-PPP2R5C regulates IL-1β ubiquitination by affecting PP2A activity in macrophages, contributing to the airway inflammation and emphysema in a murine model of COPD. PP2A and IL-1β ubiquitination in macrophages might be new therapeutic avenues for COPD therapy.
Collapse
Affiliation(s)
- Min Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Manni Zhu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyu Jia
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingjing Wu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qi Yuan
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tingting Xu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhengxia Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mao Huang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Ningfei Ji
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Mingshun Zhang
- Jiangsu Province Engineering Research Center of Antibody Drug, NHC Key Laboratory of Antibody Technique, Department of Immunology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
46
|
Dang P, Wang F, Yu H. Prognostic potential of neutrophil-to-lymphocyte ratio, platelet to lymphocyte ratio, and monocyte to lymphocyte ratio in acute myocardial infarction patients combined with chronic obstructive pulmonary disease. Front Cardiovasc Med 2024; 11:1401634. [PMID: 39070559 PMCID: PMC11272454 DOI: 10.3389/fcvm.2024.1401634] [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: 03/15/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024] Open
Abstract
Background Inflammation is considered to play an important role in chronic obstructive pulmonary disease (COPD) and acute myocardial infarction (AMI), but the relationship between inflammation and poor prognosis in these patients has not yet been studied. Methods We enrolled AMI patients combined with COPD and divided them into three groups according to the tertiles of neutrophil-to-lymphocyte ratio (NLR), platelet to lymphocyte ratio (PLR) and monocyte to lymphocyte ratio (MLR) respectively. Logistic regression analyses were used to identify risk factors for in-hospital all-cause death in these patients. Covariates were adjusted stepwise to determine the association between inflammatory markers and poor prognosis. Also, the receiver operating characteristic (ROC) curve was used to evaluate the greatest predictive indicator for all-cause death. Results A total of 281 AMI patients combined with COPD were enrolled, of which 31 experienced in-hospital mortality. The risk of all-cause death was significantly higher among those with higher NLR. The highest tertile of NLR was significantly associated with an increased risk of all-cause death (all P < 0.05). This association remained significant after adjusting for confounding factors [Odds Ratio (OR): 10.571, 95% confidence interval (CI): 2.307-48.442, P = 0.002]. Moreover, compared to MLR and PLR, NLR had the highest predictive value for all-cause death [area under the curve (AUC): 0.764, 95% CI: 0.681-0.847]. Conclusion In AMI patients combined with COPD, elevated levels of inflammation were associated with increased all-cause mortality. Compared to other inflammatory indicators, NLR may provide a more superior predictive value.
Collapse
Affiliation(s)
- Peizhu Dang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Feiyang Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Hang Yu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
47
|
Liu Y, Huang Q, Zhuang Z, Yang H, Gou X, Xu T, Liu K, Wang J, Liu B, Gao P, Cao F, Yang B, Zhang C, Chen M, Fan G. Gut virome alterations in patients with chronic obstructive pulmonary disease. Microbiol Spectr 2024; 12:e0428723. [PMID: 38785444 PMCID: PMC11218493 DOI: 10.1128/spectrum.04287-23] [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: 01/03/2024] [Accepted: 04/08/2024] [Indexed: 05/25/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the primary causes of mortality and morbidity worldwide. The gut microbiome, particularly the bacteriome, has been demonstrated to contribute to the progression of COPD. However, the influence of gut virome on the pathogenesis of COPD is rarely studied. Recent advances in viral metagenomics have enabled the rapid discovery of its remarkable role in COPD. In this study, deep metagenomics sequencing of fecal virus-like particles and bacterial 16S rRNA sequencing was performed on 92 subjects from China to characterize alterations of the gut virome in COPD. Lower richness and diversity of the gut virome were observed in the COPD subjects compared with the healthy individuals. Sixty-four viral species, including Clostridium phage, Myoviridae sp., and Synechococcus phage, showed positive relationships with pulmonary ventilation functions and had markedly declined population in COPD subjects. Multiple viral functions, mainly involved in bacterial susceptibility and the interaction between bacteriophages and bacterial hosts, were significantly declined in COPD. In addition, COPD was characterized by weakened viral-bacterial interactions compared with those in the healthy cohort. The gut virome showed diagnostic performance with an area under the curve (AUC) of 88.7%, which indicates the potential diagnostic value of the gut virome for COPD. These results suggest that gut virome may play an important role in the development of COPD. The information can provide a reference for the future investigation of diagnosis, treatment, and in-depth mechanism research of COPD. IMPORTANCE Previous studies showed that the bacteriome plays an important role in the progression of chronic obstructive pulmonary disease (COPD). However, little is known about the involvement of the gut virome in COPD. Our study explored the disease-specific virome signatures of patients with COPD. We found the diversity and compositions altered of the gut virome in COPD subjects compared with healthy individuals, especially those viral species positively correlated with pulmonary ventilation functions. Additionally, the declined bacterial susceptibility, the interaction between bacteriophages and bacterial hosts, and the weakened viral-bacterial interactions in COPD were observed. The findings also suggested the potential diagnostic value of the gut virome for COPD. The results highlight the significance of gut virome in COPD. The novel strategies for gut virome rectifications may help to restore the balance of gut microecology and represent promising therapeutics for COPD.
Collapse
Affiliation(s)
- Yue Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qingsong Huang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhenhua Zhuang
- Chengdu Life Baseline Technology Co., Ltd., Chengdu, China
| | - Hongjing Yang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoling Gou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tong Xu
- Chengdu Life Baseline Technology Co., Ltd., Chengdu, China
| | - Ke Liu
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Wang
- Department of Respiratory Medicine, Chengdu Fifth People’s Hospital, Chengdu, China
| | - Bo Liu
- Department of Respiratory Medicine, Chengdu Fifth People’s Hospital, Chengdu, China
| | - Peiyang Gao
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Feng Cao
- Chengdu Life Baseline Technology Co., Ltd., Chengdu, China
| | - Bin Yang
- Chengdu Life Baseline Technology Co., Ltd., Chengdu, China
| | - Chuantao Zhang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mei Chen
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Gang Fan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
48
|
Castro-Rodriguez JA, Turi KN, Forno E. A critical analysis of the effect of OM-85 for the prevention of recurrent respiratory tract infections or wheezing/asthma from systematic reviews with meta-analysis. Pediatr Allergy Immunol 2024; 35:e14186. [PMID: 39016384 PMCID: PMC11296687 DOI: 10.1111/pai.14186] [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: 05/21/2024] [Revised: 06/15/2024] [Accepted: 06/20/2024] [Indexed: 07/18/2024]
Abstract
Acute respiratory tract infections (RTIs) are one of the most common causes of pediatric consultations/hospitalizations and a major trigger for asthma exacerbations. Some consensus statements have recommended the use of immunostimulants to boost natural defenses against severe or repeated infections. One of the most common immunostimulants is OM-85; while several randomized clinical trials (RCTs) have evaluated its efficacy in preventing acute RTIs and wheezing/asthma exacerbations, results have been conflicting. Similarly, various systematic reviews with meta-analyses (SRMs) on OM-85 have used different strategies, populations, and outcomes; moreover, SRM conclusions are limited when the original studies are highly heterogeneous or have a low quality, hindering the generalizability of the findings. Here we summarize the evidence on the effect of OM-85 to prevent acute RTIs, wheezing/asthma episodes, or loss of asthma control in children, by including and critically evaluating all SRMs published to date. We searched for SRMs on OM-85 in three publication databases and found nine SRMs (seven for RTI, and two for wheezing/asthma). Among those, one had a high confidence evaluation of quality (AMSTAR-2 tool) and found a reduction in the total number of acute RTIs among the OM-85 group. Overall, no strong recommendations can be derived from the existing literature, mainly due to the high heterogeneity among included RCTs and SRMs. Further, large, high-quality RCTs are needed to confirm the true efficacy of OM-85 for the prevention of acute RTIs, asthma development, and asthma exacerbations.
Collapse
Affiliation(s)
- Jose A Castro-Rodriguez
- Department of Pediatric Pulmonology, School of Medicine, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Kedir N Turi
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University, Bloomington, Indiana, USA
| | - Erick Forno
- Pulmonary, Allergy/Immunology, and Sleep Medicine, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
49
|
Melo-Narváez MC, Bramey N, See F, Heinzelmann K, Ballester B, Steinchen C, Jain E, Federl K, Hu Q, Dhakad D, Behr J, Eickelberg O, Yildirim AÖ, Königshoff M, Lehmann M. Stimuli-Specific Senescence of Primary Human Lung Fibroblasts Modulates Alveolar Stem Cell Function. Cells 2024; 13:1129. [PMID: 38994981 PMCID: PMC11240317 DOI: 10.3390/cells13131129] [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: 05/15/2024] [Revised: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
Aging is the main risk factor for chronic lung diseases (CLDs) including idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD). Accordingly, hallmarks of aging like cellular senescence are increased in these patients in different lung cell types including fibroblasts. However, little is known about the different triggers that induce a senescence phenotype in different disease backgrounds and its role in CLD pathogenesis. Therefore, we characterized senescence in primary human lung fibroblasts (phLF) from control, IPF, or COPD patients at baseline and after exposure to disease-relevant insults (H2O2, bleomycin, TGF-β1) and studied their capacity to support progenitor cell potential in a lung organoid model. Bulk-RNA sequencing revealed that phLF from IPF and COPD activate different transcriptional programs but share a similar senescence phenotype at baseline. Moreover, H2O2 and bleomycin but not TGF-β1 induced senescence in phLF from different disease origins. Exposure to different triggers resulted in distinct senescence programs in phLF characterized by different SASP profiles. Finally, co-culture with bleomycin- and H2O2-treated phLF reduced the progenitor cell potential of alveolar epithelial progenitor cells. In conclusion, phLF from COPD and IPF share a conserved senescence response that varies depending on the insult and impairs alveolar epithelial progenitor capacity ex vivo.
Collapse
Affiliation(s)
- Maria Camila Melo-Narváez
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
- Institute for Lung Research, Philipps-University Marburg, German Center for Lung Research (DZL), 35043 Marburg, Germany
| | - Nora Bramey
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
| | - Fenja See
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
| | - Katharina Heinzelmann
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
| | - Beatriz Ballester
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
- Faculty of Health Sciences, Universidad Cardenal Herrera—CEU, CEU Universities, 46115 Valencia, Spain
| | - Carina Steinchen
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
| | - Eshita Jain
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
| | - Kathrin Federl
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
| | - Qianjiang Hu
- Division of Pulmonary, Allergy & Critical Care, and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (Q.H.); (O.E.); (M.K.)
| | - Deepesh Dhakad
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
| | - Jürgen Behr
- Department of Medicine V, University Hospital Munich, Medical Faculty of the LMU Munich, 81377 Munich, Germany;
| | - Oliver Eickelberg
- Division of Pulmonary, Allergy & Critical Care, and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (Q.H.); (O.E.); (M.K.)
| | - Ali Önder Yildirim
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
- Institute of Experimental Pneumology, University Hospital Munich, Ludwig-Maximilians University, 81377 Munich, Germany
| | - Melanie Königshoff
- Division of Pulmonary, Allergy & Critical Care, and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (Q.H.); (O.E.); (M.K.)
| | - Mareike Lehmann
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), German Center for Lung Research (DZL), 81377 Munich, Germany; (M.C.M.-N.); (F.S.); (C.S.); (E.J.); (D.D.); (A.Ö.Y.)
- Institute for Lung Research, Philipps-University Marburg, German Center for Lung Research (DZL), 35043 Marburg, Germany
- Lung Aging and Regeneration, Institute for Lung Health (ILH), 35392 Giessen, Germany
| |
Collapse
|
50
|
Chen Q, Zhou H, Tang J, Sun Y, Ao G, Zhao H, Chang X. An analysis of exogenous harmful substance exposure as risk factors for COPD and hypertension co-morbidity using PSM. Front Public Health 2024; 12:1414768. [PMID: 38983261 PMCID: PMC11231093 DOI: 10.3389/fpubh.2024.1414768] [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: 04/09/2024] [Accepted: 06/03/2024] [Indexed: 07/11/2024] Open
Abstract
Background Some occupational and environmental exposures could increase the risk of chronic obstructive pulmonary disease (COPD) and hypertension in various work and living environments. However, the effect of exposure to multiple exogenous harmful substances on COPD and hypertension co-morbidities remains unclear. Methods Participants were selected from eight hospitals in five provinces in China using a multistage cluster sampling procedure. Participants' demographic, exposure, and disease information were collected through questionnaires, spirometry, and blood pressure examinations. Demographic data were used as matching factors, and 1:1 matching between the exposed and non-exposed groups was performed by employing propensity score matching (PSM) to minimize the influence on the results. A one-way chi-squared analysis and multifactorial logistic regression were used to analyze the association between the exposure to exogenous harmful substances (metals and their compound dust, inorganic mineral dust, organic chemicals, and livestock by-products) and the co-morbidity of COPD and hypertension. Results There were 6,610 eligible participants in the final analysis, of whom 2,045 (30.9%) were exposed to exogenous harmful substances. The prevalence of co-morbidities of COPD and hypertension (6.0%) in the exposure group was higher than their prevalence in the total population (4.6%). After PSM, exogenous harmful substance exposure was found to be a risk factor for the co-morbidity of COPD and hypertension [odds ratio (OR) = 1.347, 95% confidence interval (CI): 1.011-1.794], which was not statistically significant before PSM (OR = 1.094, 95% CI: 0.852-1.405). Meanwhile, the results of different outcomes showed that the association between hypertension and exogenous harmful substance exposure was not statistically significant (OR = 0.965, 95% CI: 0.846-1.101). Smoking (OR = 4.702, 95% CI: 3.321-6.656), history of a respiratory disease during childhood (OR = 2.830, 95% CI: 1.600-5.006), and history of respiratory symptoms (OR = 1.897, 95% CI: 1.331-2.704) were also identified as risk factors for the co-morbidity of COPD and hypertension. Conclusion The distribution of exogenous harmful substance exposure varies in the population, and the prevalence of co-morbidities is generally higher in susceptible populations. Exposure to exogenous harmful substances was found to be a key risk factor after adjusting for demographic confounders.
Collapse
Affiliation(s)
- Qingyang Chen
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Haodong Zhou
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Jiarong Tang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Yingbiao Sun
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Geriletu Ao
- Department of Pulmonary and Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Hongjun Zhao
- Department of Pulmonary and Critical Care Medicine, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|