1
|
Chen X, Wu C, Tang F, Zhou J, Mo L, Li Y, He J. The Immune Microenvironment: New Therapeutic Implications in Organ Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e05067. [PMID: 40391706 DOI: 10.1002/advs.202505067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/28/2025] [Indexed: 05/22/2025]
Abstract
Fibrosis, characterized by abnormal deposition of structural proteins, is a major cause of tissue dysfunction in chronic diseases. The disease burden associated with progressive fibrosis is substantial, and currently approved drugs are unable to effectively reverse it. Immune cells are increasingly recognized as crucial regulators in the pathological process of fibrosis by releasing effector molecules, such as cytokines, chemokines, extracellular vesicles, metabolites, proteases, or intercellular contact. Therefore, targeting the immune microenvironment can be a potential strategy for fibrosis reduction and reversion. This review summarizes the recent advances in the understanding of the immune microenvironment in fibrosis including phenotypic and functional transformations of immune cells and the interaction of immune cells with other cells. The novel opportunities for the discovery and development of drugs for immune microenvironment remodeling and their associated challenges are also discussed.
Collapse
Affiliation(s)
- Xiangqi Chen
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuan Wu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fei Tang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jingyue Zhou
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Mo
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanping Li
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinhan He
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
2
|
Mara G, Nini G, Cotoraci C. Chronic Obstructive Pulmonary Disease and COVID-19: The Impact of Hematological Biomarkers on Disease Severity and Outcomes. J Clin Med 2025; 14:2765. [PMID: 40283596 PMCID: PMC12027599 DOI: 10.3390/jcm14082765] [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: 03/25/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Chronic obstructive pulmonary disease (COPD) patients are at heightened risk of severe COVID-19 due to underlying respiratory impairment, systemic inflammation, and immune dysregulation. This review explores the hematological changes that occur in COPD patients with COVID-19 and their implications for disease progression, prognosis, and clinical management. Methods: We conducted a comprehensive analysis of recent peer-reviewed studies from medical databases including Clarivate Analytics, PubMed, and Google Scholar. Results: Hematological alterations, such as lymphopenia, elevated neutrophil-to-lymphocyte ratio (NLR), increased D-dimer and fibrinogen levels, inflammatory anemia, and erythrocyte dysfunction, are commonly observed in COPD patients with COVID-19. These changes are linked to immune suppression, hyperinflammation, oxidative stress, and thromboembolic complications. Conclusions: Hematological biomarkers are valuable tools for early risk assessments and guiding treatment strategies in this high-risk population. The regular monitoring of D-dimer, fibrinogen, and NLR is advisable. Prophylactic anticoagulation and immunomodulatory therapies, such as corticosteroids and IL-6 and IL-1 inhibitors, may improve clinical outcomes. Further clinical studies are needed to validate personalized approaches and explore antioxidant-based interventions.
Collapse
Affiliation(s)
- Gabriela Mara
- Multidisciplinary Doctoral School, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
- Pneumology Department, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
| | - Gheorghe Nini
- Pneumology Department, Vasile Goldis Western University of Arad, 310414 Arad, Romania;
| | - Coralia Cotoraci
- Clinical Hematology Department, Vasile Goldis Western University of Arad, 310025 Arad, Romania
| |
Collapse
|
3
|
Li J, Wu X, Fu Y, Liu J, Liu Y, Li J, Qing B, Zhang Y, Gao J, He X, Wang J, Li G. Transcriptomic and metabolomic insights into neutrophil activity in COPD complicated by metabolic syndrome. Biomed Eng Online 2025; 24:43. [PMID: 40234868 PMCID: PMC11998468 DOI: 10.1186/s12938-025-01378-5] [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: 06/15/2024] [Accepted: 04/02/2025] [Indexed: 04/17/2025] Open
Abstract
OBJECTIVES Chronic obstructive pulmonary disease (COPD) frequently coexists with metabolic syndrome (MS), compounding its impact on patients' health and quality of life. This study aimed to elucidate the immune and metabolic response characteristics in COPD patients with and without MS. METHODS A total of 11,315 COPD patients admitted to the Department of Respiratory and Critical Care Medicine at the Third People's Hospital of Chengdu between January 1, 2013, and May 1, 2023, were selected. Multivariate logistic regression was conducted to identify the risk factors for acute exacerbation of chronic obstructive pulmonary disease. Moreover, from this cohort, 30 patients (18 with COPD and 12 with COPD-MS) were recruited for a further study to investigate the underlying mechanisms of COPD and COPD-MS. Blood samples were collected from these participants to perform transcriptomic and metabolomic analyses, aiming to explore the differences in immune responses and metabolic alterations between the two groups. RESULTS Our findings indicate a significant enhancement of neutrophil-mediated immune responses in COPD-MS patients. Transcriptomic analysis revealed 327 differentially expressed genes (DEGs) significantly involved in neutrophil-mediated immunity. Key metabolic pathways were disrupted, with 39 differential metabolites identified. Notably, metabolites, such as L-homoarginine and diethanolamine, which were elevated in COPD-MS patients, showed strong correlations with DEGs involved in neutrophil pathways and immune checkpoint regulation. The study also found decreased levels of IL4 and IL5RA in COPD-MS patients, suggesting a shift from Th2 to Th1 inflammatory responses, potentially contributing to glucocorticoid resistance. CONCLUSIONS COPD patients with metabolic syndrome exhibit a heightened neutrophil-mediated inflammatory response and significant metabolic disturbances, which underscores the need for precise therapeutic strategies targeting both metabolic and inflammatory pathways to improve patient outcomes and manage COPD-MS complexities effectively.
Collapse
Affiliation(s)
- Juan Li
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, China
- Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, China
| | - Xue Wu
- Clinical Medicine Department, North Sichuan Medical College, Nanchong, China
- Department of Respiratory and Critical Care Medicine, Hospital of Shimian County, Ya'an, China
| | - Yufen Fu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Respiratory and Critical Care Medicine, Longchang People's Hospital, Neijiang, China
| | - Jiliu Liu
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, China
- Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, China
| | - Yao Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Jiahuan Li
- Clinical Medicine Department, North Sichuan Medical College, Nanchong, China
| | - Bomiao Qing
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, China
- Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, China
| | - Yi Zhang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, China
- Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, China
| | - Jie Gao
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, China
- Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, China
| | - Xiang He
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, China.
- Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, China.
| | - Junyi Wang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, China.
- Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, China.
- Clinical Medicine Department, North Sichuan Medical College, Nanchong, China.
| | - Guoping Li
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, College of Medicine, Southwest Jiaotong University, Chengdu, China.
- Department of Respiratory Medicine, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Affiliated Hospital of ChongQing Medical University, Chengdu, China.
- Clinical Medicine Department, North Sichuan Medical College, Nanchong, China.
| |
Collapse
|
4
|
Cai Y, Chen H, Chen Z, Chen Q, Su Y, Chen F, Pan J, Yang Y, Hu Z, Li W, Liao H, Sun T, Cheng J, Chen W, Zhang B, Chen R. Nomogram model for predicting pressure injury in COPD patients using SII: a Chinese clinical study. Front Med (Lausanne) 2025; 12:1564099. [PMID: 40297157 PMCID: PMC12034554 DOI: 10.3389/fmed.2025.1564099] [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: 01/22/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
Objectives This study aims to investigate the association between the Systemic Immune-Inflammation Index (SII) and the development of pressure injuries (PI) in patients with chronic obstructive pulmonary disease (COPD). Additionally, a nomogram model based on the SII will be constructed to predict the probability of pressure injury (PI) occurrence in patients with COPD. Methods A retrospective analysis was performed on the clinical data of 844 patients with COPD who were admitted to the Affiliated Hospital of Guangdong Medical University between June 2018 and December 2019. Logistic regression analysis was employed to identify risk factors associated with the development of PI, and the Wald chi-square test was used to select variables for constructing a predictive nomogram. The performance of the nomogram was assessed, followed by internal validation. Additionally, clinical data from 452 patients with COPD admitted to the Second Affiliated Hospital of Guangdong Medical University between January 2024 and December 2024 were prospectively collected for external validation. Results A total of 844 patients with COPD were included in this study, with 590 cases in the training group and 254 cases in the internal validation group. The predictors included in the nomogram model were age, respiratory rate [Breathe (R)], duration of COPD history, Serum albumin (ALB), SII, paralysis, edema, and activities of daily living (ADL). The nomogram demonstrated strong predictive performance and calibration. The area under the curve and 95% confidence intervals were 0.77 (0.72-0.82) for the training group, 0.77 (0.70-0.85) for the internal validation group, and 0.73 (0.66-0.81) for the external validation group. Conclusion This study identified the SII, age, respiratory rate, duration of COPD history, ALB, paralysis, and ADL as independent risk factors for the development of PI in patients with COPD. A nomogram model was successfully developed based on SII and validated through both internal and external testing. The findings suggest that SII is a reliable predictor of PI development in patients with COPD, and the model demonstrates strong predictive performance.
Collapse
Affiliation(s)
- Yuli Cai
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Huimin Chen
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zhaojun Chen
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Qinghua Chen
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yihuan Su
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Feiju Chen
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Jingjing Pan
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yitian Yang
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zhongxing Hu
- The First Clinical School of Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Wenxi Li
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Huizhao Liao
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Tingting Sun
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Junfen Cheng
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Wenliang Chen
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Baozhi Zhang
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Riken Chen
- The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
5
|
Lu J, Xie H, Chen J, Ma B, He J, Chen Q, Li S. The impact of obstructive sleep apnea on early surgical site infections in elderly PLIF patients: a retrospective propensity score-matched analysis. J Orthop Surg Res 2025; 20:351. [PMID: 40205474 PMCID: PMC11980275 DOI: 10.1186/s13018-025-05777-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is a prevalent sleep disorder associated with intermittent hypoxia, oxidative stress, and systemic inflammation, posing risks for adverse postoperative outcomes, including surgical site infections (SSIs). Elderly patients undergoing posterior lumbar interbody fusion (PLIF) are particularly susceptible to SSIs due to advanced age, comorbidities, and prolonged surgical times. However, the role of OSA in increasing SSI risk among this population remains unclear. METHODS This retrospective cohort study analyzed 478 elderly PLIF patients from a single institution between May 2016 and June 2024. Of these, 113 were diagnosed with OSA. Propensity score matching (PSM) was performed to balance baseline characteristics, resulting in 83 matched pairs. SSI rates, hospital stays, and readmission rates were compared between the OSA and non-OSA groups. Subgroup analysis was conducted to evaluate the effects of continuous positive airway pressure (CPAP) or automatic positive airway pressure (APAP) therapy on postoperative outcomes. RESULTS After PSM, OSA patients demonstrated a significantly higher SSI incidence (13.3% vs. 3.6%, p = 0.04) compared to non-OSA patients. On multivariate analysis, OSA was the only factor that remained significantly associated with an increased risk of SSIs (Odds Ratio = 4.509, 95% CI: 1.283-21.504, p = 0.03). OSA patients also experienced longer hospital stays (10.1 ± 2.9 vs. 9.1 ± 2.0 days, p = 0.01) and elevated 30-day readmission rates (9.6% vs. 1.2%, p = 0.02). Subgroup analysis revealed that CPAP/APAP therapy reduced SSI incidence (3.9% vs. 17.5%, p = 0.08) and shortened hospital stays (9.1 ± 1.5 vs. 10.5 ± 3.2 days, p = 0.03) among OSA patients. CONCLUSION OSA significantly increases the risk of early SSIs and prolongs hospital stays in elderly PLIF patients. Subgroup analysis suggests that CPAP/APAP therapy may have benefits to OSA patients, though this association requires validation through prospective studies. These findings emphasize the importance of preoperative OSA screening and management to improve surgical outcomes in this high-risk population.
Collapse
Affiliation(s)
- Jiao Lu
- Department of Urological Surgery, People's Hospital of Yuechi County, Guang'an, 638300, Sichuan, People's Republic of China
| | - Haiyang Xie
- Department of Orthopaedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, People's Republic of China
| | - Jianwen Chen
- Department of Orthopaedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, People's Republic of China
| | - Bingjiang Ma
- Department of Orthopaedic and Reconstructive Surgery/Pediatric Orthopaedics, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, People's Republic of China
| | - Jiangtao He
- Department of Orthopaedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, People's Republic of China
| | - Qian Chen
- Department of Orthopaedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, People's Republic of China.
| | - Shuliang Li
- Department of Orthopaedic and Reconstructive Surgery/Pediatric Orthopaedics, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, People's Republic of China.
| |
Collapse
|
6
|
Yang Q, Shen X, Luo Y, Li R, Meng X, Xu P, Liu X, Bian D, Wang J, Shi J, Chen J. ELANE enhances KEAP1 protein stability and reduces NRF2-mediated ferroptosis inhibition in metabolic dysfunction-associated fatty liver disease. Cell Death Dis 2025; 16:266. [PMID: 40204709 PMCID: PMC11982220 DOI: 10.1038/s41419-025-07603-2] [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: 11/09/2024] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/11/2025]
Abstract
Neutrophil elastase (Elane) is upregulated in metabolic-associated fatty liver disease (MAFLD) and has the capacity to promote disease progression. However, the mechanism by which Elane promotes MAFLD development remains unclear. Ferroptosis, which is an iron-dependent nonapoptotic form of cell death characterized by the iron-induced accumulation of lipid reactive oxygen species (ROS), has been recently considered as an important mechanism for the development of MAFLD. In this study, we used mice of Elane-knockout (Elane-KO) and wild-type (WT), and their primary mouse hepatocytes to establish MAFLD models in vivo and vitro for elucidating the role of Elane in ferroptosis of hepatocytes and MAFLD development. Elane-KO in vivo reduced high-fat diet (HFD) induced hepatic lipid peroxidation levels and the proportion of hepatocyte death, upregulated the expression of Nrf2 and Gpx4, and downregulated Keap1 expression. Treatment with recombinant Elane increased the lipid peroxidation level of hepatocytes, increased the ferroptosis rate of hepatocytes, upregulated the expression of Keap1, enhanced the ubiquitination of Nrf2, and downregulated the expression of Nrf2 and Gpx4 in an FFA-induced MAFLD in vitro model. However, primary hepatocytes from Elane-KO mice presented opposite changes. Furthermore, an in vitro experiment revealed that Elane enhanced the protein stability of Keap1 and thus increased Keap1 expression in hepatocytes by inhibiting the lysosomal degradation of the Keap1 protein. Finally, in vitro Co-IP experiments revealed that Elane increased the protein stability of Keap1 by weakening the binding between P62 and Keap1 and ultimately promoted hepatocyte Nrf2 ubiquitination and ferroptosis in MAFLD. In conclusion, our results suggested that Elane promoted hepatocyte ferroptosis in MAFLD through the P62-Keap1-Nrf2-Gpx4 axis. Elane promotes ferroptosis in hepatocytes from fatty livers. Elane reduces the binding of P62 to Keap1, thereby increasing Keap1 protein stability and subsequently inhibiting the Nrf2/Gpx4 pathway, ultimately leading to ferroptosis in hepatocytes.
Collapse
Affiliation(s)
- Qingqing Yang
- Department of Gastroenterology, The First People's Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, China
| | - Xuan Shen
- College of Basic Medicine, Jiangsu Medical college, Yancheng, Jiangsu, China
| | - Yan Luo
- Department of Liver Diseases, Hangzhou Normal University Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Rongqing Li
- College of Clinical Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiangrui Meng
- Department of Nuclear Medicine, Xinxiang Central Hospital, The Fourth Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Ping Xu
- Department of Gastroenterology, The First People's Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, China
| | - Xuan Liu
- Department of Gastroenterology, The First People's Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, China
| | - Dongxue Bian
- Department of Gastroenterology, Yancheng TCM Hospital Affiliated with Nanjing University of Chinese Medicine, Yancheng, Jiangsu, China
| | - Jianhua Wang
- Department of Gastroenterology, The First People's Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, China.
| | - Junping Shi
- Department of Liver Diseases, Hangzhou Normal University Affiliated Hospital, Hangzhou, Zhejiang, China.
| | - Jin Chen
- Department of Gastroenterology, The First People's Hospital of Yancheng, The Yancheng Clinical College of Xuzhou Medical University, Yancheng, Jiangsu, China.
| |
Collapse
|
7
|
Xiao Y, Ma R, Liu C, Wei H, Zhou B, Gui C, Yang B, Cao Y, Zhang K, Luo B. Insights into Pulmonary Heart Disease at High Altitude: Hypoxia and PM 2.5 Interactions. Am J Respir Crit Care Med 2025; 211:660-663. [PMID: 39836437 DOI: 10.1164/rccm.202409-1802rl] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025] Open
Affiliation(s)
- Ya Xiao
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Runxue Ma
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Ce Liu
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Huanhuan Wei
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Baofeng Zhou
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Chunyan Gui
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Bo Yang
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Yunshan Cao
- Heart, Lung and Vessels Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; and
| | - Kai Zhang
- Department of Environmental Health Sciences, School of Public Health, University at Albany, State University of New York, Rensselaer, New York
| | - Bin Luo
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
8
|
Wang P, Wang J, Ge L, Gao B, Wang S, Jiang S. Automatically titrating oxygen system versus constant flow oxygen system during exercise in patients with COPD: a systematic review and meta-analysis. BMC Pulm Med 2025; 25:140. [PMID: 40155894 PMCID: PMC11951597 DOI: 10.1186/s12890-025-03594-0] [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: 12/30/2024] [Accepted: 03/11/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Hypoxemia is a common symptom among patients with chronic obstructive pulmonary disease (COPD). The constant flow oxygen system (CFOS) is often insufficient to correct this symptom. The automatically titrating oxygen system (ATOS), a new oxygen therapy mode, remains undetermined in its ability to improve exercise performance more effectively than CFOS in COPD patients. The main objective of this meta-analysis was to explore this issue. METHODS We conducted a thorough search of randomized controlled trials (RCTs) in PubMed, Embase, Web of Science (from inception to 1 November 2024). Study selection, data extraction, and risk of bias assessment were performed independently by two authors. Data synthesis was conducted using Stata software (Version 17.0). The Grading of Recommendations, Assessment, Development and Evaluation (GRADE) system was utilized to rate evidence quality. RESULTS Five eligible studies (n = 120) were included. Compared to CFOS, ATOS was more effective in extending the distance (MD = 180.28 m, 95%CI:133.03 to 227.52) and duration (MD = 237.63 s, 95%CI: 181.18 to 294.07) of endurance shuttle walking test (ESWT). Besides, ATOS could better prolong the percentage time of sustaining targeted SpO2 (92%-96%) (MD = 29.43%,95%CI:21.15 to 37.71) and relieve dyspnea at isotime (MD = -1.65, 95%CI -3.19 to -0.11) during ESWT. DISCUSSION ATOS may have more advantages in improving exercise tolerance, sustaining targeted SpO2, and ameliorating dyspnea during exercise in COPD patients. CLINICAL TRIAL REGISTRATION The review was registered with PROSPERO (The website is https://www.crd.york.ac.uk/prosp ero/, and the ID is CRD 42024574955) and we didn't make a protocol.
Collapse
Affiliation(s)
- Peijian Wang
- Department of Rehabilitation Medicine, China-Japan Friendship Hospital, 2 Yinghuayuan East St. Hepingli, Chaoyang District, Beijing, 100029, China
| | - Jing Wang
- Department of Radiotherapy, Xuzhou Central Hospital, Xuzhou, JiangSu Province, China
| | - Lijun Ge
- Department of Rehabilitation Medicine, China-Japan Friendship Hospital, 2 Yinghuayuan East St. Hepingli, Chaoyang District, Beijing, 100029, China
| | - Beiyao Gao
- Department of Rehabilitation Medicine, China-Japan Friendship Hospital, 2 Yinghuayuan East St. Hepingli, Chaoyang District, Beijing, 100029, China
| | - Siyuan Wang
- Department of Rehabilitation Medicine, China-Japan Friendship Hospital, 2 Yinghuayuan East St. Hepingli, Chaoyang District, Beijing, 100029, China.
| | - Shan Jiang
- Department of Rehabilitation Medicine, China-Japan Friendship Hospital, 2 Yinghuayuan East St. Hepingli, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
9
|
Parvin S, Bagheri H, Halabian R, Arabfard M, Ghazvini A, Vahedi E, Najafi A, Ghanei M. Comprehensive transcriptomics analysis of peripheral blood mononuclear cells in exposure to mustard gas. Int Immunopharmacol 2025; 150:114197. [PMID: 39946765 DOI: 10.1016/j.intimp.2025.114197] [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: 12/25/2024] [Revised: 01/20/2025] [Accepted: 01/28/2025] [Indexed: 03/03/2025]
Abstract
INTRODUCTION Sulfur mustard (SM) is a substance that causes blisters and has been repeatedly used by Iraq in chemical warfare against more than 100,000 Iranians. The main issue for these people is various pulmonary problems similar to chronic obstructive pulmonary disease (COPD). MATERIALS AND METHODS Our study analyzed the total RNA profile extracted using the RNA-seq technique from peripheral blood mononuclear cells (PBMCs) isolated from Mustard Lung (ML) patients of all three groups (Severe, Moderate, and Mild) in terms of disease in healthy control (HC) subjects on the BGISEQ platform (Paired-end, 7 GB data, and rRNA depletion). However, given the severe group's importance in clinical problems, we prioritized studying this group. Differentially expressed genes (DEGs) of the severe group versus HC were obtained using the limma package. DEGs were analyzed through bioinformatics tools, and their gene ontology (GO) and enrichment analysis (EA) were evaluated. Then, String-db and Cytoscape tools were used to search for the most important functional genes. RESULTS We identified SERPINA1, MAPK3, MMP9, FOXO3, SLC4A1, FCGR3B, CXCR2, PTGS2, HBA2, GPX1, IL1RN, IFNG, RPS29, CXCL1, FPR1, and RPS9 genes using hub and bottleneck criteria. Based on the analysis of important genes, several biological pathways were identified, including innate immunity, inflammatory response, and activation of neutrophils, cellular response to cytokines, and cellular response to oxidative stress, lipoxygenase pathway, and macrophage differentiation. CONCLUSION Innate immunity and neutrophils play a crucial role in the pathogenesis of these individuals. The signaling pathways of interleukins 4, 10, and 13 stimulate the differentiation of lung macrophages (MQs) into M2, essential for repair, remodeling, and inflammation. Additionally, reactive oxygen species (ROS) activate Protein kinase B (PKB), also known as AKT, through Phosphoinositide 3-kinases (PI3K) and increase the activity of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), which results in decreased histone deacetylase 2 (HDAC2) being one of the important pathways of pathophysiology in these patients.
Collapse
Affiliation(s)
- Shahram Parvin
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hasan Bagheri
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Raheleh Halabian
- Applied Microbiology Research Center, Biomedicine Technologies Institute Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Masoud Arabfard
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Ghazvini
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ensieh Vahedi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Najafi
- Molecular Biology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Liegeois MA, Hsieh A, Al-Fouadi M, Charbit AR, Yang CX, Hackett TL, Fahy JV. Cellular and molecular features of asthma mucus plugs provide clues about their formation and persistence. J Clin Invest 2025; 135:e186889. [PMID: 40091838 PMCID: PMC11910225 DOI: 10.1172/jci186889] [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: 09/10/2024] [Accepted: 01/24/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUNDMucus plugs form in acute asthma and persist in chronic disease. Although eosinophils are implicated in mechanisms of mucus pathology, many mechanistic details about mucus plug formation and persistence in asthma are unknown.METHODSUsing histology and spatial, single-cell proteomics, we characterized mucus-plugged airways from nontransplantable donor lungs of 14 patients with asthma (9 with fatal asthma and 5 with nonfatal asthma) and individuals acting as controls (10 with chronic obstructive pulmonary disease and 14 free of lung disease). Additionally, we used an airway epithelial cell-eosinophil (AEC-eosinophil) coculture model to explore how AEC mucus affects eosinophil degranulation.RESULTSAsthma mucus plugs were tethered to airways showing infiltration with innate lymphoid type 2 cells and hyperplasia of smooth muscle cells and MUC5AC-expressing goblet cells. Asthma mucus plugs were infiltrated with immune cells that were mostly dual positive for eosinophil peroxidase (EPX) and neutrophil elastase, suggesting that neutrophils internalize EPX from degranulating eosinophils. Indeed, eosinophils exposed to mucus from IL-13-activated AECs underwent CD11b- and glycan-dependent cytolytic degranulation. Dual-positive granulocytes varied in frequency in mucus plugs. Whereas paucigranulocytic plugs were MUC5AC rich, granulocytic plugs had a mix of MUC5AC, MUC5B, and extracellular DNA traps. Paucigranulocytic plugs occurred more frequently in (acute) fatal asthma and granulocytic plugs predominated in (chronic) nonfatal asthma.CONCLUSIONTogether, our data suggest that mucin-rich mucus plugs in fatal asthma form because of acute goblet cell degranulation in remodeled airways and that granulocytic mucus plugs in chronic asthma persist because of a sustaining niche characterized by epithelial cell-mucin-granulocyte cross-talk.FUNDINGNIH grants HL080414, HL107202, and AI077439.
Collapse
Affiliation(s)
- Maude A. Liegeois
- Cardiovascular Research Institute, UCSF, San Francisco, California, USA
| | - Aileen Hsieh
- Centre for Heart Lung Innovation, Vancouver, British Columbia, Canada
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - May Al-Fouadi
- Centre for Heart Lung Innovation, Vancouver, British Columbia, Canada
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Chen Xi Yang
- Centre for Heart Lung Innovation, Vancouver, British Columbia, Canada
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tillie-Louise Hackett
- Centre for Heart Lung Innovation, Vancouver, British Columbia, Canada
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - John V. Fahy
- Cardiovascular Research Institute, UCSF, San Francisco, California, USA
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, UCSF, San Francisco, California, USA
| |
Collapse
|
11
|
Liu Y, Zhao W, Hu C, Zhang Y, Qu Y. Predictive Value of the Neutrophil-to-Lymphocyte Ratio/Serum Albumin for All-Cause Mortality in Critically Ill Patients Suffering from COPD. Int J Chron Obstruct Pulmon Dis 2025; 20:659-683. [PMID: 40098661 PMCID: PMC11911821 DOI: 10.2147/copd.s497829] [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: 10/08/2024] [Accepted: 02/23/2025] [Indexed: 03/19/2025] Open
Abstract
Background Among critically ill patients, chronic obstructive pulmonary disease (COPD) is an independent risk factor for death. Recently, biomarkers such as neutrophil-lymphocyte ratio (NLR) and albumin (ALB) have been used to predict the prognosis in patients with COPD. However, the association between NLR/ALB and all-cause mortality in critically ill COPD patients remains unclear. This study aims to explore the association between the NLR/ALB and prognosis in critically ill patients with COPD. Methods Data was sourced from the Medical Information Mart for Intensive Care IV (MIMIC-IV) database. Primary outcome was 28-day all-cause mortality, with secondary outcomes being in-hospital and 90-day all-cause mortality. The area under the receiver operating characteristic curve (AUROC) was calculated to compare prognostic accuracy of NLR, NLR/ALB, PLR, SII and MLR variables. After identifying the most predictive factor, KM survival curves, Cox models and subgroup analyses were used to examine NLR/ALB's relationship with mortality in critically ill COPD patients. Additionally, patients with COPD from the National Health and Nutrition Examination Survey data (1999-2018) was used with Cox regression to investigate NLR/ALB's correlation with all-cause mortality in COPD patients. Results 1916 critically ill COPD patients from MIMIC IV, divided into quartiles by NLR/ALB levels: Q1 (NLR/ALB<1.108), Q2 (2.095>NLR/ALB≥1.108), Q3 (4.221>NLR/ALB≥2.095), Q4 (NLR/ALB≥4.221). In multivariate Cox regression, Q4 vs Q1: 28-day mortality HR=2.27 (95% CI: 1.63-3.16); 90-day mortality HR=2.06 (95% CI: 1.56-2.71); in-hospital mortality HR=1.93 (95% CI: 1.35-2.77); P<0.001. Subgroup analyses showed that the correlation between NLR/ALB and 28-day mortality was stable Additionally, we recruited 2,003 COPD patients from the NHANES that found NLR/ALB also correlated with all-cause mortality in COPD (In multivariate Cox regression: Q4 vs Q1 hR=1.92 (95% CI: 1.45-2.55, P<0.001)). Conclusion Elevated NLR/ALB levels are associated with increased all-cause mortality in critically ill patients with COPD.
Collapse
Affiliation(s)
- Yongli Liu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong Key Laboratory of Infectious Respiratory Diseases, Jinan, People’s Republic of China
| | - Wei Zhao
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong Key Laboratory of Infectious Respiratory Diseases, Jinan, People’s Republic of China
| | - Chenyang Hu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong Key Laboratory of Infectious Respiratory Diseases, Jinan, People’s Republic of China
| | - Yuxin Zhang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong Key Laboratory of Infectious Respiratory Diseases, Jinan, People’s Republic of China
| | - Yiqing Qu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Shandong Key Laboratory of Infectious Respiratory Diseases, Jinan, People’s Republic of China
| |
Collapse
|
12
|
Jia S, Chen Q, Huang W, Wang P, Zeng Y. Relationship between systemic immune response index (SIRI) and COPD: a cross-sectional study based on NHANES 2007-2012. Sci Rep 2025; 15:7887. [PMID: 40050308 PMCID: PMC11885421 DOI: 10.1038/s41598-025-90947-8] [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: 12/22/2024] [Accepted: 02/17/2025] [Indexed: 03/09/2025] Open
Abstract
Although the link between inflammation and chronic obstructive pulmonary disease (COPD) is increasingly recognized, the correlation between systemic immune response index (SIRI), a novel marker of inflammation, and COPD is unknown. This cross-sectional study used data from patients with complete lung function in NHANES 2007-2012 to explore the relationship between SIRI and COPD. We performed a series of statistical analyses on a total of 5056 participants, including multiple linear regression, smoothed curve fitting, ROC curve analysis, and subgroup analysis. In the fully corrected model, the logistic multiple regression showed that SIRI was associated with a high risk of COPD (OR1.350, 95% CI:1.220,1.493). The ROC curve showed that SIRI (AUC = 0.596) was significantly more efficient than other inflammatory factors in predicting COPD. Smoothed curve fit effect and threshold effect analyses showed a linear correlation between SIRI COPD prevalence, and subgroup analyses showed that the effect of SIRI on COPD was more pronounced in still smokers (OR 1.58, 95% CI: 1.34, 1.86) versus men (OR 1.62, 95% CI: 1.44, 1.83). The results of the interaction test provide evidence supporting SIRI as an independent risk factor for COPD.
Collapse
Affiliation(s)
- Shengqi Jia
- Department of Respiratory and Critical Care Medicine, Liyuan Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuying Chen
- Department of Respiratory and Critical Care Medicine, Liyuan Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijia Huang
- Department of Geriatrics, Liyuan Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Wang
- Department of Respiratory and Critical Care Medicine, Liyuan Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yulan Zeng
- Department of Respiratory and Critical Care Medicine, Liyuan Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
13
|
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 2025; 83:657-668. [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] [MESH Headings] [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
|
14
|
Tojo K, Yazawa T. A high fraction of inspired oxygen does not mitigate atelectasis-induced lung tissue hypoxia or injury in experimental acute respiratory distress syndrome. Sci Rep 2025; 15:1906. [PMID: 39809828 PMCID: PMC11733249 DOI: 10.1038/s41598-024-83992-2] [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: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Although alveolar hyperoxia exacerbates lung injury, clinical studies have failed to demonstrate the beneficial effects of lowering the fraction of inspired oxygen (FIO2) in patients with acute respiratory distress syndrome (ARDS). Atelectasis, which is commonly observed in ARDS, not only leads to hypoxemia but also contributes to lung injury through hypoxia-induced alveolar tissue inflammation. Therefore, it is possible that excessively low FIO2 may enhance hypoxia-induced inflammation in atelectasis, and raising FIO2 to an appropriate level may be a reasonable strategy for its mitigation. In this study, we investigated the effects of different FIO2 levels on alveolar tissue hypoxia and injury in a mechanically ventilated rat model of experimental ARDS with atelectasis. Rats were intratracheally injected with lipopolysaccharide (LPS) to establish an ARDS model. They were allocated to the low, moderate, and high FIO2 groups with FIO2 of 30, 60, and 100%, respectively, a day after LPS injection. All groups were mechanically ventilated with an 8 mL/kg tidal volume and zero end-expiratory pressure to induce dorsal atelectatic regions. Arterial blood gas analysis was performed every 2 h. After six hours of mechanical ventilation, the rats were euthanized, and blood, bronchoalveolar lavage fluid, and lung tissues were collected and analyzed. Another set of animals was used for pimonidazole staining of the lung tissues to detect the hypoxic region. Lung mechanics, ratios of partial pressure of arterial oxygen (PaO2) to FIO2, and partial pressure of arterial carbon dioxide were not significantly different among the three groups, although PaO2 changed with FIO2. The dorsal lung tissues were positively stained with pimonidazole regardless of FIO2, and the HIF-1α concentrations were not significantly different among the three groups, indicating that raising FIO2 could not rescue alveolar tissue hypoxia. Moreover, changes in FIO2 did not significantly affect lung injury or inflammation. In contrast, hypoxemia observed in the low FIO2 group caused injury to organs other than the lungs. Raising FIO2 levels did not attenuate tissue hypoxia, inflammation, or injury in the atelectatic lung region in experimental ARDS. Our results indicate that raising FIO2 levels to attenuate atelectasis-induced lung injury cannot be rationalized.
Collapse
Affiliation(s)
- Kentaro Tojo
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University School of Medicine and Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan.
| | - Takuya Yazawa
- Department of Pathology, Dokkyo Medical University School of Medicine and Graduate School of Medicine, 880 Kitakobayashi, Mibu, Shimotsugagun, Tochigi, 321-0293, Japan
| |
Collapse
|
15
|
Jiang P, Huang H, Liu Z, Xiang G, Wu X, Hao S, Li S. STAT6 deficiency mitigates the severity of pulmonary arterial hypertension caused by chronic intermittent hypoxia by suppressing Th2-inducing cytokines. Respir Res 2025; 26:13. [PMID: 39806384 PMCID: PMC11731530 DOI: 10.1186/s12931-024-03062-z] [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: 09/15/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is frequently associated with increased incidence and mortality of pulmonary hypertension (PH). The immune response contributes to pulmonary artery remodeling and OSA-related diseases. The immunologic factors linked to OSA-induced PH are not well understood. STAT6 is crucial in the signaling pathway that modulates immune response. However, the status of phosphorylated STAT6 (p-STAT6) in an OSA-induced PH mouse model remains largely unexplored. METHODS Chronic intermittent hypoxia (CIH) plays a crucial role in the progression of OSA. This study utilized a in vivo CIH model to examine the role of STAT6 in CIH-induced PH. RESULTS CIH mice exhibited pulmonary artery remodeling and pulmonary hypertension, indicated by increased right ventricular systolic pressure (RVSP), higher right ventricular to left ventricular plus septum (RV/LV + S) ratios, and significant morphological alterations compared to normoxic (Nor) mice. Increased p-STAT6 in the lungs and elevated p-STAT6 + IL-4 + producing T cells in CIH mice. STAT6 deficiency (STAT6-/-) improved PH and PA remodeling in CIH-induced PH mouse models.STAT6 deficiency impaired the T helper 2 (Th2) immune response, affecting IL-4 and IL-13 secretion. IL-4, rather than IL-13, activated STAT6 in human pulmonary artery smooth muscle cells (hPASMCs). STAT6 knockdown decreased the proliferation in IL-4 treated hPASMCs. CONCLUSION These findings exhibit the critical role of STAT6 in the pathogenesis of CIH induced PH by regulating Th2 immune response.STAT6 could be a significant therapeutic target for OSA-related PH.
Collapse
Affiliation(s)
- Pan Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- The Nutrition Department at Zhongshan Hospital, Fudan University, Shanghai, China
- The Nutrition Department, QingPu District Central Hospital, Shanghai, 200032, China
| | - Huai Huang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zilong Liu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Guiling Xiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- The Critical Care Medicine Department at Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaodan Wu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Clinical Center for Sleep Breathing Disorder and Snoring, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Shengyu Hao
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- The Critical Care Medicine Department at Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Shanqun Li
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Clinical Center for Sleep Breathing Disorder and Snoring, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
16
|
Tourn J, Crescence L, Bruzzese L, Panicot-Dubois L, Dubois C. Cellular and Molecular Mechanisms Leading to Air Travel-Induced Thrombosis. Circ Res 2025; 136:115-134. [PMID: 39745986 DOI: 10.1161/circresaha.124.325208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Venous thromboembolism, characterized by deep vein thrombosis and pulmonary embolism, is the third cardiovascular disease in the world. Deep vein thrombosis occurs when a blood clot forms in areas of impaired blood flow, and it is significantly affected by environmental factors. Local hypoxia, caused by venous stasis, plays a critical role in deep vein thrombosis under normal conditions, and this effect is intensified when the Po2 decreases, such as during air travel or high-altitude exposure. The lower oxygen levels and reduced pressure at high altitudes further contribute to deep vein thrombosis development. These conditions increase the pro-coagulant activity of neutrophils, platelets, and red blood cells, which interact on the surface of activated endothelial cells, promoting clot formation. Understanding the mechanisms involved in thrombus formation when Po2 is reduced, with or without pressure reduction, is crucial for preventing the development of venous thromboembolisms in such conditions and identifying innovative therapeutic targets. This literature review explores the mechanisms involved in thrombus formation related to high-altitude conditions and discusses the pro-coagulant consequences induced by environmental disturbances.
Collapse
Affiliation(s)
- Julie Tourn
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
| | - Lydie Crescence
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
- Plateforme Aix Marseille, Plateforme d'Imagerie Vasculaire et de Microscopie Intravitale, C2VN, Marseille, France (L.C., L.B., L.P.-D., C.D.)
| | - Laurie Bruzzese
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
- Plateforme Aix Marseille, Plateforme d'Imagerie Vasculaire et de Microscopie Intravitale, C2VN, Marseille, France (L.C., L.B., L.P.-D., C.D.)
| | - Laurence Panicot-Dubois
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
- Plateforme Aix Marseille, Plateforme d'Imagerie Vasculaire et de Microscopie Intravitale, C2VN, Marseille, France (L.C., L.B., L.P.-D., C.D.)
| | - Christophe Dubois
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
- Plateforme Aix Marseille, Plateforme d'Imagerie Vasculaire et de Microscopie Intravitale, C2VN, Marseille, France (L.C., L.B., L.P.-D., C.D.)
| |
Collapse
|
17
|
Sun CY, Tesfaigzi Y, Lee GY, Chen YH, Weiss ST, Ma KSK. Clinical effectiveness and safety of dupilumab in patients with chronic obstructive pulmonary disease: A 7-year population-based cohort study. J Allergy Clin Immunol 2025; 155:219-222.e1. [PMID: 39368553 DOI: 10.1016/j.jaci.2024.09.019] [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: 07/06/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Previous randomized controlled trials have established the efficacy of dupilumab among patients with chronic obstructive pulmonary disease (COPD) treated with triple therapy over 52 weeks of follow-up. OBJECTIVE This population-based cohort study aimed to explore the long-term safety and effectiveness of dupilumab in patients with COPD. METHODS The study included US patients with COPD who were seen between April 2017 and August 2024. Patients initiating dupilumab and therapies that incorporated long-acting β2-agonist (LABA) inhalers were included. Patients with asthma or lung cancer were excluded. The risk of outcomes occurring after initiation of dupilumab versus LABA-containing therapies was measured. For detailed methods, please see the Methods section in this article's Online Repository at www.jacionline.org. RESULTS A total of 1521 dupilumab initiators and 1521 propensity score-matched patients who were receiving LABA-based therapies were included. Receiving dupilumab was associated with lower all-cause mortality (hazard ratio [HR] = 0.53, 95% CI = 0.43-0.65), fewer emergency department visits (HR = 0.78, 95% CI = 0.69-0.89), and lower acute exacerbation rates (HR = 0.59, 95% CI = 0.53-0.65). Dupilumab was also associated with reductions in the requirement for short-acting β2-agonists (HR = 0.48, 95% CI = 0.43-0.52) and short-acting muscarinic antagonists (HR = 0.43, 95% CI = 0.37-0.49) for symptom control. Additionally, dupilumab decreased rates of subsequent pneumonia (HR = 0.65, 95% CI = 0.50-0.86), and COPD-relevant comorbidities, including new-onset heart failure (HR = 0.69, 95% CI = 0.53-0.90) and new-onset anxiety (HR = 0.70, 95% CI =0.53-0.93). CONCLUSIONS In patients with COPD, dupilumab was associated with a lower mortality rate, fewer emergency department visits, and a reduced risk of acute exacerbations, respiratory symptoms, and respiratory infections. More studies are needed to validate the efficacy of dupilumab among patients with COPD of various severities.
Collapse
Affiliation(s)
- Chuan-Yen Sun
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa; Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yohannes Tesfaigzi
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Gin-Yi Lee
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Yi-Hsuan Chen
- Department of Environmental Health and Engineering, Johns Hopkins University, Baltimore, Md
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Kevin Sheng-Kai Ma
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa.
| |
Collapse
|
18
|
Zhao B, Zhao Y, Sun X. Mechanism and therapeutic targets of circulating immune cells in diabetic retinopathy. Pharmacol Res 2024; 210:107505. [PMID: 39547465 DOI: 10.1016/j.phrs.2024.107505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Diabetic retinopathy (DR) continues to be the leading cause of preventable vision loss among working-aged adults, marked by immune dysregulation within the retinal microenvironment. Typically, the retina is considered as an immune-privileged organ, where circulating immune cells are restricted from entry under normal conditions. However, during the progression of DR, this immune privilege is compromised as circulating immune cells breach the barrier and infiltrate the retina. Increasing evidence suggests that vascular and neuronal degeneration in DR is largely driven by the infiltration of immune cells, particularly neutrophils, monocyte-derived macrophages, and lymphocytes. This review delves into the mechanisms and therapeutic targets associated with these immune cell populations in DR, offering a promising and innovative approach to managing the disease.
Collapse
Affiliation(s)
- Bowen Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yin Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Xufang Sun
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
19
|
Nisar A, Khan S, Li W, Hu L, Samarawickrama PN, Gold NM, Zi M, Mehmood SA, Miao J, He Y. Hypoxia and aging: molecular mechanisms, diseases, and therapeutic targets. MedComm (Beijing) 2024; 5:e786. [PMID: 39415849 PMCID: PMC11480526 DOI: 10.1002/mco2.786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024] Open
Abstract
Aging is a complex biological process characterized by the gradual decline of cellular functions, increased susceptibility to diseases, and impaired stress responses. Hypoxia, defined as reduced oxygen availability, is a critical factor that influences aging through molecular pathways involving hypoxia-inducible factors (HIFs), oxidative stress, inflammation, and epigenetic modifications. This review explores the interconnected roles of hypoxia in aging, highlighting how hypoxic conditions exacerbate cellular damage, promote senescence, and contribute to age-related pathologies, including cardiovascular diseases, neurodegenerative disorders, cancer, metabolic dysfunctions, and pulmonary conditions. By examining the molecular mechanisms linking hypoxia to aging, we identify key pathways that serve as potential therapeutic targets. Emerging interventions such as HIF modulators, antioxidants, senolytics, and lifestyle modifications hold promise in mitigating the adverse effects of hypoxia on aging tissues. However, challenges such as the heterogeneity of aging, lack of reliable biomarkers, and safety concerns regarding hypoxia-targeted therapies remain. This review emphasizes the need for personalized approaches and advanced technologies to develop effective antiaging interventions. By integrating current knowledge, this review provides a comprehensive framework that underscores the importance of targeting hypoxia-induced pathways to enhance healthy aging and reduce the burden of age-related diseases.
Collapse
Affiliation(s)
- Ayesha Nisar
- Key Laboratory of Genetic Evolution & Animal Models, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingChina
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
| | - Sawar Khan
- Department of Cell Biology, School of Life SciencesCentral South UniversityChangshaHunanChina
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
| | - Wen Li
- Department of EndocrinologyThe Second Affiliated Hospital of Dali University (the Third People's Hospital of Yunnan Province)KunmingYunnanChina
| | - Li Hu
- Key Laboratory of Genetic Evolution & Animal Models, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingChina
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
| | - Priyadarshani Nadeeshika Samarawickrama
- Key Laboratory of Genetic Evolution & Animal Models, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingChina
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
| | - Naheemat Modupeola Gold
- Key Laboratory of Genetic Evolution & Animal Models, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingChina
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
| | - Meiting Zi
- Key Laboratory of Genetic Evolution & Animal Models, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
| | | | - Jiarong Miao
- Department of GastroenterologyThe First Affiliated Hospital of Kunming Medical UniversityKunmingYunnanChina
| | - Yonghan He
- Key Laboratory of Genetic Evolution & Animal Models, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
- Kunming College of Life ScienceUniversity of Chinese Academy of SciencesKunmingChina
- Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of ZoologyChinese Academy of SciencesKunmingYunnanChina
| |
Collapse
|
20
|
Wang D, Wang M, Sun S, Zhang C, Song Y, Li J, Song B, Lv H, Wang S, Jiang W. Hypoxia-induced NLRP3 inflammasome activation via the HIF-1α/NF-κB signaling pathway in human dental pulp fibroblasts. BMC Oral Health 2024; 24:1156. [PMID: 39343901 PMCID: PMC11441079 DOI: 10.1186/s12903-024-04936-w] [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: 03/19/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Previous studies have reported the link between hypoxic conditions and NLRP3 inflammasome-mediated pulpal inflammation in the progression of pulpitis. However, the underlying mechanism has not been fully elucidated. This study aimed to investigate the role of HIF-1α in the regulation of NLRP3 inflammasome pathway via NF-κB signaling under hypoxic conditions with or without LPS in human dental pulp fibroblasts (HDPFs) during the progression of pulpitis. METHODS HIF-1α plasmids or siRNAs were used to upregulate or downregulate HIF-1α in HDPFs, respectively. The effect of hypoxia with or without LPS on the NF-κB signaling and NLRP3 inflammasome pathway was analyzed by immunofluorescence staining, qRT-PCR, western blotting and ELISA. RESULTS The hypoxic conditions alone induced ASC oligomerization and NLRP3/CASP1 inflammasome pathway activation via NF-κB signaling in a time-dependent manner in HDPFs. The upregulation of HIF-1α further promoted hypoxia-induced ASC oligomerization and NLRP3/CASP1 inflammasome pathway activation via NF-κB signaling compared to the hypoxia-induced group. In comparison, downregulation of HIF-1α inhibited ASC oligomerization and NLRP3/CASP1 inflammasome pathway activation via NF-κB signaling compared to the hypoxia-induced group. Additionally, LPS plus hypoxia further promoted HIF-1α expression and NLRP3/ASC/CASP1 inflammasome pathway activation via NF-κB signaling compared to the hypoxia-induced group. CONCLUSIONS HIF-1α served as a positive regulator of NLRP3/ASC/CASP1 inflammasome pathway activation via NF-κB signaling in HDPFs in the sterile pulpal inflammation and caries-related pulpitis microenvironment. The finding of a novel functional HIF-1α-NF-κB-NLRP3 axis provides insight into the link between the hypoxic microenvironment and pulpal inflammation, thus supporting a promising therapeutic strategy for the control of pulpal inflammation.
Collapse
Affiliation(s)
- Diya Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Minghao Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Shukai Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Pediatric Dentistry, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chongyang Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Ya Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Jianing Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
| | - Bing Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, CF14 4XY, UK
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Haipeng Lv
- Department of Stomatology, Xi'an Daxing Hospital, Xi'an, Shaanxi, China.
| | - Shengchao Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China.
| | - Wenkai Jiang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry & Endodontics, School of Stomatology, Fourth Military Medical University, No.145 Western Changle Road, Xi'an, Shaanxi, 710032, China.
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, CF14 4XY, UK.
| |
Collapse
|
21
|
Chen M, Chang S, Xu Y, Guo H, Liu J. Dietary Beetroot Juice - Effects in Patients with COPD: A Review. Int J Chron Obstruct Pulmon Dis 2024; 19:1755-1765. [PMID: 39099609 PMCID: PMC11296515 DOI: 10.2147/copd.s473397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/20/2024] [Indexed: 08/06/2024] Open
Abstract
Chronic Obstructive Pulmonary Disease (COPD) exerts a severe toll on human health and the economy, with high prevalence and mortality rates. The search for bioactive components effective in the treatment of COPD has become a focal point of research. Beetroot juice, readily accessible and cost-effective, is noted for its ability to enhance athletic performance and for its preventive and therapeutic impact on hypertension. Beetroot juice is a rich source of dietary nitrates and modulates physiological processes via the nitrate-nitrite- nitric oxide pathway, exerting multiple beneficial effects such as antihypertensive, bronchodilatory, anti-inflammatory, antioxidant, hypoglycemic, and lipid-lowering actions. This paper provides a review of the existing research on the effects of beetroot juice on COPD, summarizing its potential in enhancing exercise capacity, lowering blood pressure, improving vascular function, and ameliorating sleep quality among patients with COPD. The review serves as a reference for the prospective use of beetroot juice in the symptomatic improvement of COPD, as well as in the prevention of exacerbations and associated comorbidities.
Collapse
Affiliation(s)
- Mingming Chen
- The First Clinical Medical College of Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| | - Shuting Chang
- The First Clinical Medical College of Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| | - Yunpeng Xu
- The First Clinical Medical College of Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
| | - Hong Guo
- Department of Critical Care Medicine, Gansu Provincial Maternity and Child-Care Hospital, Lanzhou City, Gansu Province, People’s Republic of China
| | - Jian Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou City, Gansu Province, People’s Republic of China
- Gansu Provincial Maternity and Child-Care Hospital (Gansu Provincial Center Hospital), Lanzhou City, Gansu Province, People’s Republic of China
| |
Collapse
|
22
|
Lisk C, Fan A, Cendali FI, Kakiuchi K, Swindle D, Pak DI, Tolson R, Grier A, Buesing K, Zaeske S, D’Alessandro A, Borden MA, Irwin DC. Peritoneal Infusion of Oxygen Microbubbles Alters the Metabolomic Profile of the Lung and Spleen in Acute Hypoxic Exposure. Bioengineering (Basel) 2024; 11:761. [PMID: 39199719 PMCID: PMC11352204 DOI: 10.3390/bioengineering11080761] [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: 06/11/2024] [Revised: 07/10/2024] [Accepted: 07/17/2024] [Indexed: 09/01/2024] Open
Abstract
Administration of oxygen microbubbles (OMBs) has been shown to increase oxygen and decrease carbon dioxide in systemic circulation, as well as reduce lung inflammation and promote survival in preclinical models of hypoxia caused by lung injury. However, their impact on microenvironmental oxygenation remains unexplored. Herein, we investigated the effects of intraperitoneal administration of OMBs in anesthetized rats exposed to hypoxic ventilation (FiO2 = 0.14). Blood oxygenation and hemodynamics were evaluated over a 2 h time frame, and then organ and tissue samples were collected for hypoxic and metabolic analyses. Data showed that OMBs improved blood SaO2 (~14%) and alleviated tissue hypoxia within the microenvironment of the kidney and intestine at 2 h of hypoxia. Metabolomic analysis revealed OMBs induced metabolic differences in the cecum, liver, kidney, heart, red blood cells and plasma. Within the spleen and lung, principal component analysis showed a metabolic phenotype more comparable to the normoxic group than the hypoxic group. In the spleen, this shift was characterized by reduced levels of fatty acids and 2-hydroxygluterate, alongside increased expression of antioxidant enzymes such as glutathione and hypoxanthine. Interestingly, there was also a shuttle effect within the metabolism of the spleen from the tricarboxylic acid cycle to the glycolysis and pentose phosphate pathways. In the lung, metabolomic analysis revealed upregulation of phosphatidylethanolamine and phosphatidylcholine synthesis, indicating a potential indirect mechanism through which OMB administration may improve lung surfactant secretion and prevent alveolar collapse. In addition, cell-protective purine salvage was increased within the lung. In summary, oxygenation with intraperitoneal OMBs improves systemic blood and local tissue oxygenation, thereby shifting metabolomic profiles of the lung and spleen toward a healthier normoxic state.
Collapse
Affiliation(s)
- Christina Lisk
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver—Anschutz Medical Campus, Denver, CO 80204, USA; (C.L.); (D.S.); (D.I.P.); (R.T.); (S.Z.)
| | - Alex Fan
- Biomedical Engineering Program, University of Colorado, Boulder, CO 80309, USA; (A.F.); (K.K.); (M.A.B.)
| | - Francesca I. Cendali
- Department of Biochemistry & Molecular Genetics, Graduate School, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (F.I.C.); (A.G.); (A.D.)
| | - Kenta Kakiuchi
- Biomedical Engineering Program, University of Colorado, Boulder, CO 80309, USA; (A.F.); (K.K.); (M.A.B.)
| | - Delaney Swindle
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver—Anschutz Medical Campus, Denver, CO 80204, USA; (C.L.); (D.S.); (D.I.P.); (R.T.); (S.Z.)
| | - David I. Pak
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver—Anschutz Medical Campus, Denver, CO 80204, USA; (C.L.); (D.S.); (D.I.P.); (R.T.); (S.Z.)
| | - Robert Tolson
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver—Anschutz Medical Campus, Denver, CO 80204, USA; (C.L.); (D.S.); (D.I.P.); (R.T.); (S.Z.)
| | - Abby Grier
- Department of Biochemistry & Molecular Genetics, Graduate School, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (F.I.C.); (A.G.); (A.D.)
| | - Keely Buesing
- Department of Surgery, Division of Acute Care Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Seth Zaeske
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver—Anschutz Medical Campus, Denver, CO 80204, USA; (C.L.); (D.S.); (D.I.P.); (R.T.); (S.Z.)
| | - Angelo D’Alessandro
- Department of Biochemistry & Molecular Genetics, Graduate School, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (F.I.C.); (A.G.); (A.D.)
| | - Mark A. Borden
- Biomedical Engineering Program, University of Colorado, Boulder, CO 80309, USA; (A.F.); (K.K.); (M.A.B.)
- Department of Mechanical Engineering, University of Colorado, Boulder, CO 80309, USA
| | - David C. Irwin
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver—Anschutz Medical Campus, Denver, CO 80204, USA; (C.L.); (D.S.); (D.I.P.); (R.T.); (S.Z.)
| |
Collapse
|
23
|
Li R, Li H, Wang X, Peng Y. Protective Effects of Velvet Antler Methanol Extracts on Hypoxia-Induced Damage in Caenorhabditis elegans through HIF-1 and ECH-8 Mediated Lipid Accumulation. Nutrients 2024; 16:2257. [PMID: 39064700 PMCID: PMC11280314 DOI: 10.3390/nu16142257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/06/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Velvet antler, a traditional tonic widely used in East Asia for its health benefits, is explored in this study for its protective effects against hypoxia-induced damage using Caenorhabditis elegans (C. elegans) as a model. Hypoxia, characterized by low oxygen availability, induces significant physiological stress and potential tissue damage. Our research demonstrates that methanol extracts from velvet antler (MEs) enhance the survival of C. elegans under hypoxic conditions. This enhancement is achieved through the stabilization of hypoxia-inducible factor-1 (HIF-1) and the promotion of lipid accumulation, both of which are crucial for mitigating cellular damage. Specifically, MEs improve mitochondrial function, increase ATP production, and aid in the recovery of physical activity in C. elegans post-hypoxia or following hypoxia-reoxygenation (HR). The pivotal role of HIF-1 is underscored by the loss of these protective effects when HIF-1 function is inhibited. Additionally, our findings reveal that the gene related to lipid metabolism, ech-8, significantly contributes to the lipid accumulation that enhances resilience to hypoxia in C. elegans treated with MEs. These results not only highlight the therapeutic potential of velvet antler in modern medical applications, particularly for conditions involving hypoxic stress, but also provide insights into the molecular mechanisms by which MEs confer protection against hypoxic damage.
Collapse
Affiliation(s)
- Ru Li
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China;
| | - Hongyuan Li
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China;
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China;
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Yinghua Peng
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China;
| |
Collapse
|
24
|
Du D, Zhang G, Xu D, Liu L, Hu X, Zeng T, Shen Y, Luo F. Association between systemic inflammatory markers and chronic obstructive pulmonary disease: A population-based study. Heliyon 2024; 10:e31524. [PMID: 38818179 PMCID: PMC11137537 DOI: 10.1016/j.heliyon.2024.e31524] [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: 10/21/2023] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 06/01/2024] Open
Abstract
Objective To investigate whether inflammatory indices, including the systemic immune-inflammation index (SII), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), product of platelet and neutrophil count (PPN), and lymphocyte-to-monocyte ratio (LMR), correlate with chronic obstructive pulmonary disease (COPD). Methods This was a cross-sectional study from the National Health and Nutrition Examination Survey (NHANES) database 2007-2018. The SII, NLR, PLR, PPN and LMR were calculated based on blood cell counts and were log2-transformed. COPD was diagnosed via a questionnaire or spirometry examination. Multivariate logistic regression, sensitivity analysis, subgroup analyses, and interaction tests were performed to evaluate the relationships. Results 23,875 participants, including 1000 COPD patients (453 diagnosed via spirometry examination, 547 diagnosed via a questionnaire), were enrolled in this study. Positive associations were observed between SII (OR 1.231, 95 % CI 1.081,1.401), NLR (OR 1.223, 95 % CI 1.064,1.405), PLR (OR 1.325, 95 % CI 1.086,1.617), PPN (OR 1.157, 95 % CI 1.031,1.298) and COPD, while a negative association was obtained between LMR and COPD (OR 0.794, 95 % CI 0.666,0.948) after covariate adjustments. When divided COPD patients into spirometry-based and questionnaire-based, only SII (OR 1.310, 95%CI 1.122,1.529), PLR (OR 1.669, 95%CI 1.272,2.191) and PPN (OR 1.218, 95%CI 1.050,1.412) significantly correlated with spirometry-based COPD, while only NLR (OR 1.303, 95%CI 1.055,1.609) and LMR (OR 0.524, 95%CI 0.406,0.677) significantly correlated with questionnaire-based COPD after covariate adjustments. Conclusion Significant associations are observed between different inflammation indices and COPD. Heterogeneity exists between spirometry-based and questionnaire-based COPD patients. Future studies are needed to verify the results.
Collapse
Affiliation(s)
- Dongru Du
- Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, Chengdu, 610041, China
- West China School of Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Guangyue Zhang
- West China School of Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Dan Xu
- Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, Chengdu, 610041, China
| | - Lian Liu
- Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, Chengdu, 610041, China
| | - Xueru Hu
- Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, Chengdu, 610041, China
| | - Tingting Zeng
- Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, Chengdu, 610041, China
| | - Yongchun Shen
- Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, Chengdu, 610041, China
| | - Fengming Luo
- Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University and Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, Chengdu, 610041, China
| |
Collapse
|
25
|
Cheetham CJ, McKelvey MC, McAuley DF, Taggart CC. Neutrophil-Derived Proteases in Lung Inflammation: Old Players and New Prospects. Int J Mol Sci 2024; 25:5492. [PMID: 38791530 PMCID: PMC11122108 DOI: 10.3390/ijms25105492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Neutrophil-derived proteases are critical to the pathology of many inflammatory lung diseases, both chronic and acute. These abundant enzymes play roles in key neutrophil functions, such as neutrophil extracellular trap formation and reactive oxygen species release. They may also be released, inducing tissue damage and loss of tissue function. Historically, the neutrophil serine proteases (NSPs) have been the main subject of neutrophil protease research. Despite highly promising cell-based and animal model work, clinical trials involving the inhibition of NSPs have shown mixed results in lung disease patients. As such, the cutting edge of neutrophil-derived protease research has shifted to proteases that have had little-to-no research in neutrophils to date. These include the cysteine and serine cathepsins, the metzincins and the calpains, among others. This review aims to outline the previous work carried out on NSPs, including the shortcomings of some of the inhibitor-orientated clinical trials. Our growing understanding of other proteases involved in neutrophil function and neutrophilic lung inflammation will then be discussed. Additionally, the potential of targeting these more obscure neutrophil proteases will be highlighted, as they may represent new targets for inhibitor-based treatments of neutrophil-mediated lung inflammation.
Collapse
Affiliation(s)
- Coby J. Cheetham
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| | - Michael C. McKelvey
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| | - Daniel F. McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK;
| | - Clifford C. Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| |
Collapse
|
26
|
Pang X, Liu X. Immune Dysregulation in Chronic Obstructive Pulmonary Disease. Immunol Invest 2024; 53:652-694. [PMID: 38573590 DOI: 10.1080/08820139.2024.2334296] [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] [Indexed: 04/05/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a disease whose incidence increase with age and is characterised by chronic inflammation and significant immune dysregulation. Inhalation of toxic substances cause oxidative stress in the lung tissue as well as airway inflammation, under the recruitment of chemokines, immune cells gathered and are activated to play a defensive role. However, persistent inflammation damages the immune system and leads to immune dysregulation, which is mainly manifested in the reduction of the body's immune response to antigens, and immune cells function are impaired, further destroy the respiratory defensive system, leading to recurrent lower respiratory infections and progressive exacerbation of the disease, thus immune dysregulation play an important role in the pathogenesis of COPD. This review summarizes the changes of innate and adaptive immune-related cells during the pathogenesis of COPD, aiming to control COPD airway inflammation and improve lung tissue remodelling by regulating immune dysregulation, for further reducing the risk of COPD progression and opening new avenues of therapeutic intervention in COPD.
Collapse
Affiliation(s)
- Xichen Pang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaoju Liu
- Department of Gerontal Respiratory Medicine, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
27
|
Zheng H, Wang G, Wang Y, Wang Q, Sun T. Combined analysis of bulk RNA and single-cell RNA sequencing to identify pyroptosis-related markers and the role of dendritic cells in chronic obstructive pulmonary disease. Heliyon 2024; 10:e27808. [PMID: 38509896 PMCID: PMC10950670 DOI: 10.1016/j.heliyon.2024.e27808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 02/23/2024] [Accepted: 03/06/2024] [Indexed: 03/22/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by dyspnea caused by airflow limitation. Further development may lead to decreased lung function and other lung diseases. Pyroptosis is a type of programmed cell death that involves multiple pathways. For example, the pathway induced by the NLR family pyrin domain containing 3 (NLRP3) inflammasome is closely associated with COPD exacerbation. Therefore, in this study, various machine learning algorithms were applied to screen for diagnostically relevant pyroptosis-related genes from the GEO dataset, and the results were verified using external datasets. The results showed that deep neural networks and logistic regression algorithms had the highest AUC of 0.91 and 0.74 in the internal and external test sets, respectively. Here, we explored the immune landscape of COPD using diagnosis-related genes. We found that the infiltrating abundance of dendritic cells significantly differed between the COPD and control groups. Finally, the communication patterns of each cell type were explored based on scRNA-seq data. The critical role of significant pathways involved in communication between DCS and other cell populations in the occurrence and progression of COPD was identified.
Collapse
Affiliation(s)
- Huiyan Zheng
- Department of Health Management Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Guifeng Wang
- Department of Health Management Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yunlai Wang
- Department of Health Management Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qixian Wang
- Department of Health Management Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ting Sun
- Department of Health Management Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
28
|
Jin T, Liu X, Wang Y, Qi Y, Li X, Wang L, He X. Network pharmacology prediction, molecular docking and in vitro experiment explored the potential mechanism of Gaoyuan'an capsule in improving hypoxia tolerance. THE PHARMACOGENOMICS JOURNAL 2024; 24:8. [PMID: 38485921 DOI: 10.1038/s41397-024-00327-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Tibetan medicine Gaoyuan'an capsule (GYAC) is widely used to prevent pulmonary edema at high altitude, but the specific mechanism has not been explored. In this study, we analyzed the mechanism of GYAC in hypoxia tolerance, and provided a new idea for the prevention and treatment of altitude disease. METHODS The effective components and corresponding targets of GYAC were screened out by the Chinese herbal medicine network database, and the key targets of hypoxia tolerance were retrieved by Genecards, OMIM and PubMed database. Cytoscape 3.7.2 was used to construct GYAC ingredient-target-hypoxia tolerance-related target network. GO function annotation and KEGG enrichment analysis were performed to predict the pathways in which target genes may be involved, and molecular docking was used to verify the binding ability of the compound to target genes. In vitro, the above results were further verified by molecular experiment. RESULTS We found that GYAC can improve hypoxia tolerance by regulating various target genes, including IL6, IFNG, etc. The main regulatory pathways were HIF-1 signaling pathway. Molecular docking showed that the affinity between luteolin and target genes (IL6, IFNG) were better. In vitro, we observed that hypoxia can inhibit cell viability and promote apoptosis of H9C2 cell. And hypoxia can promote the expression of LDH. After the addition of luteolin, the decrease of cell viability, the increase of cell apoptosis, LDH release and the decrease of mitochondrial membrane potential were inhibited. Besides, inflammatory related factors (IL-6, IL-10, IL-2, IFNG and VEGFA) expression were also inhibited hypoxic cell models. CONCLUSIONS The results of network pharmacology and molecular docking showed that luteolin, a monomeric component of GYAC, played a role in hypoxia tolerance through a variety of target genes, such as IL6, IFNG. What's more, we have discovered that luteolin can reduce the inflammatory response in cardiac myocytes, thereby alleviating mitochondrial damage, and ultimately enhancing the hypoxia tolerance of H9C2 cardiomyocytes.
Collapse
Affiliation(s)
- Tianbo Jin
- School of Basic Medical Sciences, Xizang Minzu University, Xianyang, Shaanxi, 712082, China
| | - Xiaoli Liu
- School of Basic Medical Sciences, Xizang Minzu University, Xianyang, Shaanxi, 712082, China
| | - Yuhe Wang
- Department of Clinical Laboratory, the Affiliated Hospital of Xizang Minzu University, Xianyang, Shaanxi, 712082, China
| | - Yijin Qi
- School of Basic Medical Sciences, Xizang Minzu University, Xianyang, Shaanxi, 712082, China
| | - Xuemei Li
- School of Basic Medical Sciences, Xizang Minzu University, Xianyang, Shaanxi, 712082, China
| | - Li Wang
- School of Basic Medical Sciences, Xizang Minzu University, Xianyang, Shaanxi, 712082, China
| | - Xue He
- School of Basic Medical Sciences, Xizang Minzu University, Xianyang, Shaanxi, 712082, China.
| |
Collapse
|
29
|
Ye H, Yang Q, Guo H, Wang X, Cheng L, Han B, Hong M, Ma F, Li M, Wu X, Chen F, Zhu J, Chen S, Zheng S, Li J. Internalisation of neutrophils extracellular traps by macrophages aggravate rheumatoid arthritis via Rab5a. RMD Open 2024; 10:e003847. [PMID: 38485453 PMCID: PMC10941157 DOI: 10.1136/rmdopen-2023-003847] [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: 10/25/2023] [Accepted: 02/22/2024] [Indexed: 03/17/2024] Open
Abstract
OBJECTIVES Although elevated levels of neutrophil extracellular traps (NETs) have been reported in patients with rheumatoid arthritis (RA), the role of NETs in RA and the relationship between NETs and macrophages in the pathogenesis of RA requires further research. Here, we sought to determine the role of NETs in RA pathogenesis and reveal the potential mechanism. METHODS Neutrophil elastase (NE) and myeloperoxidase (MPO)-DNA were measured in human serum and synovium. NETs inhibitor GSK484 was used to examine whether NETs involved with RA progression. We stimulated macrophages with NETs and detected internalisation-related proteins to investigate whether NETs entry into macrophages and induced inflammatory cytokines secretion through internalisation. To reveal mechanisms mediating NETs-induced inflammation aggravation, we silenced GTPases involved in internalisation and inflammatory pathways in vivo and in vitro and detected downstream inflammatory pathways. RESULTS Serum and synovium from patients with RA showed a significant increase in NE and MPO, which positively correlated to disease activity. Inhibiting NETs formation alleviated the collagen-induced arthritis severity. In vitro, NETs are internalised by macrophages and located in early endosomes. Rab 5a was identified as the key mediator of the NETs internalisation and inflammatory cytokines secretion. Rab 5a knockout mice exhibited arthritis alleviation. Moreover, we found that NE contained in NETs activated the Rab5a-nuclear factor kappa B (NF-κB) signal pathway and promoted the inflammatory cytokines secretion in macrophages. CONCLUSIONS This study demonstrated that NETs-induced macrophages inflammation to aggravate RA in Rab 5a dependent manner. Mechanically, Rab5a mediated internalisation of NETs by macrophages and NE contained in NETs promoted macrophages inflammatory cytokines secretion through NF-κB-light-chain-enhancer of activated B cells signal pathway. Therapeutic targeting Rab 5a or NE might extend novel strategies to minimise inflammation in RA.
Collapse
Affiliation(s)
- Haixin Ye
- Department of Rheumatology and Immunology, Nanfang Hospital,Southern Medical University, Guangzhou, Guangdong, China
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Qian Yang
- Department of Rheumatology and Immunology, Nanfang Hospital,Southern Medical University, Guangzhou, Guangdong, China
| | - Huaxia Guo
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xing Wang
- Department of Rheumatology and Immunology, Nanfang Hospital,Southern Medical University, Guangzhou, Guangdong, China
| | - Lifang Cheng
- Department of Rheumatology and Immunology, Nanfang Hospital,Southern Medical University, Guangzhou, Guangdong, China
| | - Bingqi Han
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Mukeng Hong
- Department of Rheumatology and Immunology, Nanfang Hospital,Southern Medical University, Guangzhou, Guangdong, China
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Fopei Ma
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Meng Li
- Department of Rheumatology and Immunology, Nanfang Hospital,Southern Medical University, Guangzhou, Guangdong, China
| | - Xianghui Wu
- Laboratory Animal Research Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Feilong Chen
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Junqing Zhu
- Department of Rheumatology and Immunology, Nanfang Hospital,Southern Medical University, Guangzhou, Guangdong, China
| | - Shixian Chen
- Department of Rheumatology and Immunology, Nanfang Hospital,Southern Medical University, Guangzhou, Guangdong, China
| | - Songyuan Zheng
- Department of Rheumatology and Immunology, Nanfang Hospital,Southern Medical University, Guangzhou, Guangdong, China
| | - Juan Li
- Department of Rheumatology and Immunology, Nanfang Hospital,Southern Medical University, Guangzhou, Guangdong, China
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
30
|
Maeda T, Dransfield MT. Chronic obstructive pulmonary disease and cardiovascular disease: mechanistic links and implications for practice. Curr Opin Pulm Med 2024; 30:141-149. [PMID: 38085609 PMCID: PMC10948016 DOI: 10.1097/mcp.0000000000001040] [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] [Indexed: 01/25/2024]
Abstract
PURPOSE OF REVIEW Chronic obstructive pulmonary disease (COPD) and cardiovascular disease (CVD) are both significant burdens on the healthcare system and often coexist. Mechanistic links between the two conditions and their clinical impact are increasingly understood. RECENT FINDINGS Recent studies demonstrate multiple mechanisms by which the pathobiology of COPD may have negative effects on the cardiovascular system. These include extrapulmonary consequences of the COPD inflammatory state, cardiac autonomic dysfunction, which has been recently implicated in worsening respiratory symptoms and exacerbation risk, and mechanical effects of lung hyperinflation on left ventricular diastolic function.Clinical studies have consistently shown a high prevalence of CVD in COPD patients and worsened outcomes (and vice versa ). Exacerbations of COPD have also been demonstrated to dramatically increase the risk of cardiovascular events. While some safety concerns exist, medications for COPD and cardiovascular disease should be used in accordance with respective guidelines. However, real-world data show suboptimal management for patients with COPD and CVD. SUMMARY COPD and cardiovascular disease have complicated interrelationships. Further mechanistic studies may lead to defining better targets for interventions. Education for medical professionals and implementation of novel screening protocols should be encouraged to fill in the gaps in clinical care for these patients.
Collapse
Affiliation(s)
- Tetsuro Maeda
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
31
|
Yang J, Shen X, Qin M, Zhou P, Huang FH, You Y, Wang L, Wu JM. Suppressing inflammatory signals and apoptosis-linked sphingolipid metabolism underlies therapeutic potential of Qing-Jin-Hua-Tan decoction against chronic obstructive pulmonary disease. Heliyon 2024; 10:e24336. [PMID: 38318072 PMCID: PMC10839876 DOI: 10.1016/j.heliyon.2024.e24336] [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: 07/05/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Background Qing-Jin-Hua-Tan decoction (QJHTD) is a classic traditional Chinese medicine (TCM) prescription that first appeared in the ancient book Yi-Xue-Tong-Zhi. QJHTD has shown effectiveness for treating chronic obstructive pulmonary disease (COPD), although its mechanisms of action are still perplexing. The molecular mechanisms underlying the curative effects of QJHTD on COPD is worth exploring. Methods In vitro antiapoptotic and antiinflammatory activities of QJHTD were evaluated using cell viability, proliferation, apoptosis rate, and expression of IL-1β and TNF-α in BEAS-2B and RAW264.7 cells challenged with cigarette smoke (CS) extract (CSE) and lipopolysaccharide (LPS). In vivo therapeutic activities of QJHTD were evaluated using respiratory parameters (peak inspiratory flow (PIFb) and peak expiratory flow (PEFb) values), histopathology (mean linear intercept, MLI), and proinflammatory cytokine (IL-1β and TNF-α) and cleaved caspase-3 (c-Casp3) levels in the lung tissue of CS-LPS-exposed BALB/c mice. Network pharmacology-based prediction, transcriptomic analysis, and metabolic profiling were employed to investigate the signaling molecules and metabolites pertinent to the anti-COPD action of QJHTD. Results Increased cell viability and proliferation with decreased apoptosis rate and proinflammatory cytokine expression were noted after QJHTD intervention. QJHTD administration elevated PEFb and PIFb values, reduced MLI, and inhibited IL-1β, TNF-α, and c-Casp3 expression in vivo. Integrated network pharmacology-transcriptomics revealed that suppressing inflammatory signals (IL-1β, IL-6, TNF, IκB-NF-κB, TLR, and MAPK) and apoptosis contributed to the anti-COPD property of QJHTD. Metabolomic profiling unveiled prominent roles for the suppression of apoptosis and sphingolipid (SL) metabolism and the promotion of choline (Ch) metabolism in the anti-COPD effect of QJHTD. Integrative transcriptomics-metabolomics unraveled the correlation between SL metabolism and apoptosis. In silico molecular docking revealed that acacetin, as an active compound in QJHTD, could bind with high affinity to MEK1, MEK2, ERK1, ERK2, Bcl2, NF-κB, and alCDase target proteins. Conclusion The therapeutic effect of QJHTD on COPD is dependent on regulating inflammatory signals and apoptosis-directed SL metabolism. These findings provide deeper insights into the molecular mechanism of action of QJHTD against COPD and justify its theoretical promise in novel pharmacotherapy for this multifactorial disease.
Collapse
Affiliation(s)
- Jing Yang
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, PR China
- School of Pharmacy, Southwest Medical University, Luzhou 646000, PR China
| | - Xin Shen
- Department of Traditional Chinese Pharmacy, Chengdu First People's Hospital, Chengdu 610041, PR China
| | - Mi Qin
- School of Pharmacy, Southwest Medical University, Luzhou 646000, PR China
| | - Ping Zhou
- School of Pharmacy, Southwest Medical University, Luzhou 646000, PR China
| | - Fei-Hong Huang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, PR China
| | - Yun You
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Long Wang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, PR China
| | - Jian-Ming Wu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, PR China
| |
Collapse
|
32
|
Sun Y, Sun B, Ren Z, Xue M, Zhu C, Liu Q. Heparin-binding protein as a predictor of mortality in patients with diabetes mellitus and community-acquired pneumonia in intensive care unit : a propensity score matched study. World J Emerg Med 2024; 15:263-272. [PMID: 39050224 PMCID: PMC11265634 DOI: 10.5847/wjem.j.1920-8642.2024.033] [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: 05/29/2023] [Accepted: 11/20/2023] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Patients with diabetes mellitus (DM) are vulnerable to community-acquired pneumonia (CAP), which have a high mortality rate. We aimed to investigate the value of heparin-binding protein (HBP) as a prognostic marker of mortality in patients with DM and CAP. METHODS This retrospective study included CAP patients who were tested for HBP at intensive care unit (ICU) admission from January 2019 to April 2020. Patients were allocated to the DM or non-DM group and paired with propensity score matching. Baseline characteristics and clinical outcomes up to 90 days were evaluated. The primary outcome was the 10-day mortality. Receiver operating characteristic (ROC) curves, Kaplan-Meier analysis, and Cox regression were used for statistical analysis. RESULTS Among 152 enrolled patients, 60 pairs were successfully matched. There was no significant difference in 10-day mortality, while more patients in the DM group died within 28 d (P=0.024) and 90 d (P=0.008). In the DM group, HBP levels at ICU admission were higher in 10-day non-survivors than in 10-day survivors (median 182.21 [IQR: 55.43-300] ng/ml vs. median 66.40 [IQR: 34.13-107.85] ng/mL, P=0.019), and HBP levels could predict the 10-day mortality with an area under the ROC curve of 0.747. The cut-off value, sensitivity, and specificity were 160.6 ng/mL, 66.7%, and 90.2%, respectively. Multivariate Cox regression analysis indicated that HBP was an independent prognostic factor for 10-day (HR 7.196, 95%CI: 1.596-32.455, P=0.01), 28-day (HR 4.381, 95%CI: 1.449-13.245, P=0.009), and 90-day mortality (HR 4.581, 95%CI: 1.637-12.819, P=0.004) in patients with DM. CONCLUSION Plasma HBP at ICU admission was associated with the 10-day, 28-day, and 90-day mortality, and might be a prognostic factor in patients with DM and CAP.
Collapse
Affiliation(s)
- Yuhan Sun
- Translational Medicine Center, Department of Emergency Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Baoqing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510230, China
| | - Zhigang Ren
- Department of Infectious Disease, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Mingshan Xue
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510230, China
| | - Changju Zhu
- Henan Medical Key Laboratory of Emergency and Trauma Research, Department of Emergency Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qi Liu
- Translational Medicine Center, Department of Emergency Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
33
|
Zhang R, Zhang Y, Liu Z, Pei Y, He Y, Yu J, You C, Ma L, Fang F. Association between neutrophil-to-albumin ratio and long-term mortality of aneurysmal subarachnoid hemorrhage. BMC Neurol 2023; 23:374. [PMID: 37858065 PMCID: PMC10585913 DOI: 10.1186/s12883-023-03433-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: 11/28/2022] [Accepted: 10/10/2023] [Indexed: 10/21/2023] Open
Abstract
OBJECTIVE The prognosis of aneurysmal subarachnoid hemorrhage (aSAH) survivors is concerning. The goal of this study was to investigate and demonstrate the relationship between the neutrophil-to-albumin ratio (NAR) and long-term mortality of aSAH survivors. METHODS A retrospective observational cohort study was conducted at Sichuan University West China Hospital between January 2009 and June 2019. The investigation of relationship between NAR and long-term mortality was conducted using univariable and multivariable Cox regression models. To demonstrate the predictive performance of different biomarkers over time, time-dependent receiver operating characteristic curve (ROC) analysis and decision curve analysis (DCA) were created. RESULTS In total, 3173 aSAH patients were included in this study. There was a strong and continuous relationship between NAR levels and long-term mortality (HR 3.23 95% CI 2.75-3.79, p < 0.001). After adjustment, the result was still significant (adjusted HR 1.78 95% CI 1.49-2.12). Compared with patients with the lowest quartile (< 0.15) of NAR levels, the risk of long-term mortality in the other groups was higher (0.15-0.20: adjusted HR 1.30 95% CI 0.97-1.73; 0.20-0.28: adjusted HR 1.37 95% CI 1.03-1.82; >0.28: adjusted HR 1.74 95% CI 1.30-2.32). Results in survivors were found to be still robust. Moreover, out of all the inflammatory markers studied, NAR demonstrated the highest correlation with long-term mortality. CONCLUSIONS A high level of NAR was associated with increased long-term mortality among patients with aSAH. NAR was a promising inflammatory marker for long-term mortality of aSAH.
Collapse
Affiliation(s)
- Renjie Zhang
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yu Zhang
- Center for Evidence Based Medical and Clinical Research, Affiliated Hospital of Chengdu University, Chengdu, Sichuan, China
| | - Zheran Liu
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yiyan Pei
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yan He
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jiayi Yu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Chao You
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lu Ma
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| | - Fang Fang
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
34
|
Yang C, Yang L, Yang L, Li S, Ye L, Ye J, Chen C, Zeng Y, Zhu M, Lin X, Peng Q, Wang Y, Jin M. Plasma Proteomics Study Between the Frequent Exacerbation and Infrequent Exacerbation Phenotypes of Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2023; 18:1713-1728. [PMID: 37581107 PMCID: PMC10423573 DOI: 10.2147/copd.s408361] [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: 02/21/2023] [Accepted: 07/09/2023] [Indexed: 08/16/2023] Open
Abstract
Background Frequent exacerbation (FE) and infrequent exacerbation (IE) are two phenotypes of chronic obstructive pulmonary disease (COPD), of which FE is associated with a higher incidence of exacerbation and a serious threat to human health. Because the pathogenesis mechanisms of FE are unclear, this study aims to identify FE-related proteins in the plasma via proteomics for use as predictive, diagnostic, and therapeutic biomarkers of COPD. Methods A cross-sectional study was conducted in which plasma protein profiles were analyzed in COPD patients at stable stage, and differentially expressed proteins (DEPs) were screened out between the FE and IE patients. FE-related DEPs were identified using data-independent acquisition-based proteomics and bioinformatics analyses. In addition, FE-related candidates were verified by enzyme-linked immunosorbent assay. Results In this study, 47 DEPs were screened out between the FE and IE groups, including 20 upregulated and 27 downregulated proteins. Key biological functions (eg, neutrophil degranulation, extracellular exosome, protein homodimerization activity) and signaling pathways (eg, arginine and proline metabolism) were enriched in association with the FE phenotype. Receiver operating characteristic (ROC) analysis of the 11 combined DEPs revealed an area under the curve of 0.985 (p <0.05) for discriminating FE from IE. Moreover, correlation and ROC curve analyses indicated that creatine kinase, M-type (CKM) and fat storage-inducing transmembrane protein 1 (FITM1) might be clinically significant in patients with the FE phenotype. In addition, plasma expression levels of CKM and FITM1 were validated to be significantly decreased in the FE group compared with the IE group (CKM: p <0.01; FITM1: p <0.05). Conclusion In this study, novel insights into COPD pathogenesis were provided by investigating and comparing plasma protein profiles between the FE and IE patients. CKM, FITM1, and a combinative biomarker panel may serve as useful tools for assisting in the precision diagnosis and effective treatment of the FE phenotype of COPD.
Collapse
Affiliation(s)
- Chengyu Yang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Huadong Hospital, Fudan University, Shanghai, 200040, People’s Republic of China
| | - Li Yang
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
| | - Lei Yang
- Longhua Innovation Institute for Biotechnology, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Shuiming Li
- Longhua Innovation Institute for Biotechnology, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Ling Ye
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Jinfeng Ye
- Longhua Innovation Institute for Biotechnology, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Chengshui Chen
- Department of Pulmonary and Critical Care Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Zhejiang, 324000, People’s Republic of China
| | - Yiming Zeng
- Department of Pulmonary and Critical Care Medicine, the Second Affiliated Hospital of Fujian Medical University, Respiratory Medicine Center of Fujian Province, Quanzhou, Fujian, 362000, People’s Republic of China
| | - Mengchan Zhu
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Xiaoping Lin
- Department of Pulmonary and Critical Care Medicine, the Second Affiliated Hospital of Fujian Medical University, Respiratory Medicine Center of Fujian Province, Quanzhou, Fujian, 362000, People’s Republic of China
| | - Qing Peng
- Department of Pulmonary and Critical Care Medicine, Minhang Hospital, Fudan University, Shanghai, 201199, People’s Republic of China
| | - Yun Wang
- Longhua Innovation Institute for Biotechnology, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong, 515041, People’s Republic of China
| | - Meiling Jin
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
- Department of Allergy, Zhongshan Hospital, Fudan University, Shanghai, 200032, People’s Republic of China
| |
Collapse
|
35
|
Liang Z, Zheng X, Wang Y, Chu K, Gao Y. Using system biology and bioinformatics to identify the influences of COVID-19 co-infection with influenza virus on COPD. Funct Integr Genomics 2023; 23:175. [PMID: 37221323 DOI: 10.1007/s10142-023-01091-3] [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: 04/10/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 05/25/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has speedily increased mortality globally. Although they are risk factors for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), less is known about the common molecular mechanisms behind COVID-19, influenza virus A (IAV), and chronic obstructive pulmonary disease (COPD). This research used bioinformatics and systems biology to find possible medications for treating COVID-19, IAV, and COPD via identifying differentially expressed genes (DEGs) from gene expression datasets (GSE171110, GSE76925, GSE106986, and GSE185576). A total of 78 DEGs were subjected to functional enrichment, pathway analysis, protein-protein interaction (PPI) network construct, hub gene extraction, and other potentially relevant disorders. Then, DEGs were discovered in networks including transcription factor (TF)-gene connections, protein-drug interactions, and DEG-microRNA (miRNA) coregulatory networks by using NetworkAnalyst. The top 12 hub genes were MPO, MMP9, CD8A, HP, ELANE, CD5, CR2, PLA2G7, PIK3R1, SLAMF1, PEX3, and TNFRSF17. We found that 44 TFs-genes, as well as 118 miRNAs, are directly linked to hub genes. Additionally, we searched the Drug Signatures Database (DSigDB) and identified 10 drugs that could potentially treat COVID-19, IAV, and COPD. Therefore, we evaluated the top 12 hub genes that could be promising DEGs for targeted therapy for SARS-CoV-2 and identified several prospective medications that may benefit COPD patients with COVID-19 and IAV co-infection.
Collapse
Affiliation(s)
- Zihao Liang
- Clinical Research Center, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xudong Zheng
- Department of Immunology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuan Wang
- Clinical Research Center, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Kai Chu
- Department of Vaccine Clinical Evaluation, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China.
| | - Yanan Gao
- Department of Immunology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
36
|
Padilha C, Philp AM. A Perspective on "Hypoxia Resistance is an Inherent Phenotype of the Mouse Flexor Digitorum Brevis Skeletal Muscle". FUNCTION 2023; 4:zqad024. [PMID: 37342416 PMCID: PMC10278979 DOI: 10.1093/function/zqad024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 06/22/2023] Open
Affiliation(s)
- Camila Padilha
- Biology of Ageing Laboratory, Centenary Institute, Royal Prince Alfred Hospital, Missenden Rd, NSW 2050, Australia
- Centre for Healthy Ageing, Centenary Institute, Missenden Road, Sydney, NSW 2050, Australia
- Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia
- Faculty of Health, School of Sport, Exercise and Rehabilitation Sciences, University of Technology Sydney, Ultimo, NSW 2021, Australia
| | | |
Collapse
|
37
|
Poli V, Zanoni I. Neutrophil intrinsic and extrinsic regulation of NETosis in health and disease. Trends Microbiol 2023; 31:280-293. [PMID: 36344311 PMCID: PMC9974585 DOI: 10.1016/j.tim.2022.10.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 11/08/2022]
Abstract
Neutrophil extracellular traps (NETs) evolved to protect the host against microbial infections and are formed by a web-like structure of DNA that is decorated with antimicrobial effectors. Due to their potent inflammatory functions, NETs also cause tissue damage and can favor and/or aggravate inflammatory diseases. This multipronged activity of NETs requires that the induction, release, and degradation of NETs are tightly regulated. Here we describe the key pathways that are intrinsic to neutrophils and regulate NETosis, and we review the most recent findings on how neutrophil extrinsic factors participate in the formation of NETs. In particular, we emphasize how bystander cells contribute to modifying the capacity of neutrophils to undergo NETosis. Finally, we discuss how these neutrophil extrinsic processes can be harnessed to protect the host against the excessive inflammation elicited by uncontrolled NET release.
Collapse
Affiliation(s)
- Valentina Poli
- Harvard Medical School, Boston Children's Hospital, Division of Immunology, Division of Gastroenterology, Boston, MA, USA
| | - Ivan Zanoni
- Harvard Medical School, Boston Children's Hospital, Division of Immunology, Division of Gastroenterology, Boston, MA, USA.
| |
Collapse
|
38
|
Regard L, Burgel PR, Roche N. Inhaled therapy, cardiovascular risk and benefit-risk considerations in COPD: innocent until proven guilty, or vice versa? Eur Respir J 2023; 61:61/2/2202135. [PMID: 36758999 DOI: 10.1183/13993003.02135-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 12/06/2022] [Indexed: 02/11/2023]
Affiliation(s)
- Lucile Regard
- Université Paris Cité, Inserm U1016, Institut Cochin, Paris, France
- Respiratory Medicine, Cochin Hospital; Assistance Publique Hôpitaux de Paris Centre, Paris, France
| | - Pierre-Régis Burgel
- Université Paris Cité, Inserm U1016, Institut Cochin, Paris, France
- Respiratory Medicine, Cochin Hospital; Assistance Publique Hôpitaux de Paris Centre, Paris, France
| | - Nicolas Roche
- Université Paris Cité, Inserm U1016, Institut Cochin, Paris, France
- Respiratory Medicine, Cochin Hospital; Assistance Publique Hôpitaux de Paris Centre, Paris, France
| |
Collapse
|
39
|
Giri M, He L, Hu T, Puri A, Zheng X, Dai H, Guo S. Blood Urea Nitrogen Is Associated with In-Hospital Mortality in Critically Ill Patients with Acute Exacerbation of Chronic Obstructive Pulmonary Disease: A Propensity Score Matching Analysis. J Clin Med 2022; 11:jcm11226709. [PMID: 36431186 PMCID: PMC9699438 DOI: 10.3390/jcm11226709] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/28/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Elevated blood urea nitrogen (BUN) level is associated with a higher risk of mortality in various diseases; however, the association between BUN level and in-hospital mortality in patients with acute exacerbation of chronic obstructive pulmonary disease (AECOPD) admitted to the intensive care unit (ICU) is not known. This study aimed to investigate the relationship between BUN level and in-hospital mortality in patients with AECOPD admitted to the ICU. Methods: In this retrospective cohort study, AECOPD patients were identified from the Medical Information Mart for Intensive Care (MIMIC-IV) database. Multivariate regression was used to elucidate the relationship between BUN level and in-hospital mortality, and propensity score matching (PSM) was used to adjust confounders. Receiver operating characteristics and Kaplan−Meier curves were used to evaluate the relationship between BUN level and in-hospital mortality. Results: Data from 1201 patients were analyzed. The all-cause in-hospital mortality was 13.7%. BUN levels were significantly higher in non-survivors compared to the survival group before (p < 0.001) and after (p = 0.005) PSM. Multivariate analysis indicated that elevated BUN levels were independently associated with increased risk of in-hospital mortality both before (p = 0.002) and after (p = 0.015) PSM. The optimal BUN cut-off value for in-hospital mortality in critical patients with AECOPD before (>23 mg/dL) and after (>22 mg/dL) PSM was comparable. Compared with the low BUN group, the hazard ratio (HR) of the high BUN group was 1.8987 (before PSM) and 1.7358 (after PSM). Conclusions: Higher BUN levels were significantly associated with an increased risk of in-hospital mortality in critically ill patients with AECOPD. As a widely available and rapidly measured biomarker, BUN may be useful in the risk stratification of critically ill AECOPD patients. The results need to be verified in prospective studies.
Collapse
Affiliation(s)
- Mohan Giri
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No 1 Youyi Road, Yuzhong, Chongqing 400016, China
| | - Lin He
- Department of Respiratory and Critical Care Medicine, Chongqing University Fuling Hospital, Chongqing 408000, China
| | - Tianyang Hu
- Precision Medicine Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Anju Puri
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No 1 Youyi Road, Yuzhong, Chongqing 400016, China
| | - Xiaozhuo Zheng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No 1 Youyi Road, Yuzhong, Chongqing 400016, China
| | - Haiyun Dai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No 1 Youyi Road, Yuzhong, Chongqing 400016, China
| | - Shuliang Guo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No 1 Youyi Road, Yuzhong, Chongqing 400016, China
- Correspondence:
| |
Collapse
|
40
|
Gomez JC, Doerschuk CM. Hypoxia Can Make Neutrophils Hyper, Potentially Wreaking Havoc during Exacerbations in Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2022; 205:862-864. [PMID: 35148486 PMCID: PMC9838619 DOI: 10.1164/rccm.202201-0086ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- John C. Gomez
- Marsico Lung Institute,Division of Pulmonary Diseases and Critical Care MedicineUniversity of North CarolinaChapel Hill, North Carolina
| | - Claire M. Doerschuk
- Marsico Lung Institute,Division of Pulmonary Diseases and Critical Care MedicineUniversity of North CarolinaChapel Hill, North Carolina
| |
Collapse
|