1
|
Hayton C, Ahmed W, Terrington D, White IR, Chaudhuri N, Leonard C, Wilson AM, Fowler SJ. Relationship between exhaled volatile organic compounds and lung function change in idiopathic pulmonary fibrosis. Thorax 2025:thorax-2024-222321. [PMID: 40306949 DOI: 10.1136/thorax-2024-222321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Volatile organic compounds (VOCs) in exhaled breath have shown promise as biomarkers in idiopathic pulmonary fibrosis (IPF). We analysed breath from 57 people with IPF using thermal desorption-gas chromatography-mass spectrometry to identify VOCs related to lung function change over 12 months. A LASSO regression model selected 63 VOCs associated with relative change in forced vital capacity (8 with correlation coefficient (CC) ≥0.20 on Spearman's rank analysis), and 28 associated with relative change in diffusion capacity of the lung for carbon monoxide % predicted (12 with CC ≥0.20). Secondary analyses demonstrated a correlation between VOCs and baseline lung function parameters and association with survival. This study suggests that there may be a volatile signature of prognosis in IPF that merits further validation.
Collapse
Affiliation(s)
- Conal Hayton
- NIHR-Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, UK
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences; Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Waqar Ahmed
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences; Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | | | - Iain R White
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences; Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Laboratory for Environmental and Life Sciences, The University of Nova Gorica, Nova Gorica, Slovenia
| | - Nazia Chaudhuri
- School of Medicine, Faculty of Life and Health Sciences, University of Ulster, Londonderry, UK
- Western Healthcare Trust, Londonderry, UK
| | - Colm Leonard
- NIHR-Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Andrew M Wilson
- Norwich Medical School, University of East Anglia, Norwich, UK
- Department of Respiratory Medicine, Norfolk and Norwich NHS Foundation Trust, Norwich, UK
| | - Stephen J Fowler
- NIHR-Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, UK
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences; Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
2
|
Xie T, Zheng Y, Zhang L, Zhao J, Wu H, Li Y. PGRN knockdown alleviates pulmonary fibrosis regulating the Akt/GSK3β signaling pathway. Int Immunopharmacol 2025; 152:114443. [PMID: 40088870 DOI: 10.1016/j.intimp.2025.114443] [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: 08/27/2024] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a serious, chronic, and progressive disease with increased collagen deposition and the collapse of lung structures. Currently, the antifibrotic drugs for PF treatment, nintedanib and pirfenidone, have been proven to reduce the decline of pulmonary function in PF, but both have side effects, and to date, there is no significantly effective treatment to halt the progression of PF. The aim of this study was to investigate the molecular mechanism of pregranuloprotein (PGRN) in pulmonary fibrosis through in vitro and in vivo experiments. METHODS PF models was induced in animals using bleomycin (BLM) and treated MRC-5 cells with TGF-β1. The mRNA expression of PGRN in fasting peripheral blood samples was measured via RT-qPCR and ELSA. PGRN siRNAs were synthesized and transfected into MRC-5 cells. MAZ51, an activator of the Akt/GSK3β pathway, was applied in recovery experiment. The proliferation and apoptosis of MRC-5 cells were determined using the CCK8 kit, MTT kit, and Muse® Cell Analyzer. H&E and Masson staining were applied to evaluate the inflammatory and fibrosis in mouse lung tissue. Levels of PGRN, inflammatory factors (IL-6 and IL-1β), fibrosis markers (α-SMA, COL-I and COL-III), and Akt/GSK3β pathway-related proteins (AKT, GSK-3β and β-catenin) were determined in tissues or cells by ELISA, RT-qPCR, western blot, or Immunofluorescence. RESULTS PGRN mRNA expression was elevated in the plasma of PF patients. In TGF-β1 induced MRC-5 cells, PGRN knockdown reduced the levels of IL-6, IL-1β, α-SMA, COL-I and COL-III, and suppressed the phosphorylation of AKT and GSK-β. Treatment with MAZ51 partially reversed the effect of PGRN knockdown on TGF-β1-induced PF. Moreover, PGRN knockdown mitigated BLM-induced alveolar destruction and wall thickening, inflammatory cell infiltration, and collagen deposition in mice. It also reduced the expression of α-SMA, TGF-β1, COL-I, COL-III, β-catenin, and the phosphorylation of AKT and GSK-3β in BLM-treated mice. CONCLUSIONS PGRN knockdown alleviates PF in vitro and in vivo by modulating the Akt/GSK3β signaling pathway, proposing that PGRN could serve as a potential therapy or adjuvant therapy for lung fibrosis.
Collapse
Affiliation(s)
- Tian Xie
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China.
| | - Yamei Zheng
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Lei Zhang
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Jie Zhao
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Haihong Wu
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China.
| | - Yaqing Li
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China.
| |
Collapse
|
3
|
Lu J, Wang Z, Zhang L. Single-cell transcriptome analysis revealing mechanotransduction via the Hippo/YAP pathway in promoting fibroblast-to-myofibroblast transition and idiopathic pulmonary fibrosis development. Gene 2025; 943:149271. [PMID: 39855369 DOI: 10.1016/j.gene.2025.149271] [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: 09/27/2024] [Revised: 12/12/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
OBJECTIVE Idiopathic pulmonary fibrosis (IPF) is an irreversible and fatal interstitial lung disease, characterized by excessive extracellular matrix (ECM) secretion that disrupts normal alveolar structure. This study aims to explore the potential molecular mechanisms underlying the promotion of IPF development. METHODS Firstly, we compared the transcriptome and single-cell sequencing data from lung tissue samples of patients with IPF and healthy individuals. Subsequently, we conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses on the differentially expressed genes (DEGs). Furthermore, we employed sodium alginate hydrogels with varying degrees of crosslinking to provide differential mechanical stress, mimicking the mechanical microenvironment in vivo during lung fibrosis. On this basis, we examined cytoskeletal remodeling in fibroblasts MRC-5, mRNA expression of multiple related genes, immunofluorescence localization, and cellular proliferation capacity. RESULTS Bioinformatics analysis revealed a series of DEGs associated with IPF. Further functional and pathway enrichment analyses indicated that these DEGs were primarily enriched in ECM-related biological processes. Single-cell sequencing data revealed that fibroblasts and myofibroblasts are the main contributors to excessive ECM secretion and suggested activation of mechanotransduction and the Hippo/YAP signaling pathway in myofibroblasts. Cellular experiments demonstrated that sodium alginate hydrogels with different stiffness can simulate different mechanical stress environments, thereby affecting cytoskeletal rearrangement and Hippo/YAP pathway activity in MRC-5 lung fibroblasts. Notably, high levels of mechanical stress promoted YAP nuclear translocation, increased expression of type I collagen and α-SMA, and enhanced proliferative capacity. Additionally, we also found that fibroblasts primarily participate in mechanotransduction through the Rho/ROCK and Integrin/FAK pathways under high mechanical stress conditions, ultimately upregulating the gene expression of CCNE1/2, CTGF, and FGF1. CONCLUSION Our study uncovers the crucial role of cytoskeletal mechanotransduction in myofibroblast transformation and IPF development through activation of the Hippo/YAP pathway, providing new insights into understanding the pathogenesis of IPF.
Collapse
Affiliation(s)
- Jiaqi Lu
- Department of Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, China.
| | - Zhenhua Wang
- Department of Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, China
| | - Liguo Zhang
- Department of Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, China
| |
Collapse
|
4
|
Hu Z, Xu J, Shen R, Lin L, Su Y, Xie C, You G, Zhou Y, Huang K. Combination of Biological Aging and Genetic Susceptibility Helps Identifying At-Risk Population of Venous Thromboembolism: A Prospective Cohort Study of 394 041 Participants. Am J Hematol 2025; 100:575-583. [PMID: 39840850 DOI: 10.1002/ajh.27605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/31/2024] [Accepted: 01/13/2025] [Indexed: 01/23/2025]
Abstract
Phenotypic age acceleration (PhenoAgeAccel) is a novel clinical aging indicator. This study was carried out to investigate the relationship between PhenoAgeAccel and the incidence of VTE, as well as to integrate PhenoAgeAccel with genetic susceptibility to improve risk stratification of VTE. The study included 394 041 individuals from the UK Biobank. Phenotypic age was calculated based on actual age and clinical biomarkers. PhenoAgeAccel presents the residual obtained from a linear regression of phenotypic age against actual age, reflecting the rate of aging. Significant associations were observed between PhenoAgeAccel and higher risk of VTE (Hazard ratio [HR] 1.37, 95% CI: 1.32-1.42), deep vein thrombosis (DVT, HR 1.35, 95% CI: 1.29-1.42), and PE (pulmonary embolism, HR 1.41, 95% CI: 1.34-1.48) in the findings. PhenoAgeAccel exhibited a significant additive interaction with genetic susceptibility. Biologically older participants with high genetic risk have a 3.83 (95% CI: 3.51-4.18) folds risk of VTE, a 3.59 (95% CI: 3.21-4.03) folds risk of DVT, and 4.39 (95% CI: 3.88-4.98) folds risk of PE, in comparison to biologically younger participants with low genetic risk. Mediation analyses indicated that PhenoAgeAccel mediated approximately 6% of the association between cancer and VTE, and about 20% of the association between obesity and VTE. Our study indicated that PhenoAgeAccel is significantly associated with higher risk of VTE, and can be combined with genetic risk to improve VTE risk stratification. Additionally, PhenoAgeAccel holds promise as a clinical biomarker for guiding targeted prevention and treatment strategies for VTE.
Collapse
Affiliation(s)
- Zhensheng Hu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jiatang Xu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
| | - Runnan Shen
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Urology Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Liling Lin
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yangfan Su
- Department of Cardiovascular Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chaoyu Xie
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Department of Cardiovascular Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Guochang You
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Department of Vascular Surgery, the First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
| | - Yi Zhou
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Kai Huang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Department of Cardiovascular Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
5
|
Johannson KA, Adegunsoye A, Behr J, Cottin V, Glanville AR, Glassberg MK, Goobie GC, Jenkins RG, Kim JS, Lee CT, Redlich CA, Richeldi L, Salisbury ML, Tetley T, Corte TJ. Impact of Environmental Exposures on the Development and Progression of Fibrotic Interstitial Lung Disease. Am J Respir Crit Care Med 2025; 211:560-568. [PMID: 39745380 DOI: 10.1164/rccm.202409-1730pp] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/17/2024] [Indexed: 04/02/2025] Open
Affiliation(s)
- Kerri A Johannson
- Department of Medicine and
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | | | - Jürgen Behr
- Department of Medicine V, Ludwig Maximilian University Hospital, German Center for Lung Research, Ludwig Maximilian University Munich, Munich, Germany
| | - Vincent Cottin
- Department of Respiratory Medicine, National Reference Centre for Rare Pulmonary Diseases, European Respiratory Network- Respiratory Diseases, Louis Pradel Hospital, Lyon Public Hospitals, Lyon, France
- Mixed Research Unit 754, French National Research Institute for Agriculture, Food and Environment, Claude Bernard University Lyon 1, Lyon, France
| | - Allan R Glanville
- Macquarie Respiratory and Sleep Unit, Macquarie University, Sydney, New South Wales, Australia
| | - Marilyn K Glassberg
- Department of Medicine, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois
| | - Gillian C Goobie
- Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - R Gisli Jenkins
- National Heart and Lung Institute, National Institute for Health and Care Research Imperial Biomedical Research Centre, Imperial College London, London, United Kingdom
- Interstitial Lung Disease Centre, Royal Brompton and Harefield Hospital, London, United Kingdom
| | - John S Kim
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Cathryn T Lee
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Carrie A Redlich
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Luca Richeldi
- Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Margaret L Salisbury
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Terry Tetley
- Lung Cell Biology, Airways Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Tamera J Corte
- Department of Respiratory Medicine, Royal Prince Alfred Hospital and University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
6
|
Sack C, Wojdyla DM, MacMurdo MG, Gassett A, Kaufman JD, Raghu G, Redlich CA, Li P, Olson AL, Leonard TB, Todd JL, Neely ML, Snyder LD, Gulati M. Long-Term Air Pollution Exposure and Severity of Idiopathic Pulmonary Fibrosis: Data from the Idiopathic Pulmonary Fibrosis Prospective Outcomes (IPF-PRO) Registry. Ann Am Thorac Soc 2025; 22:378-386. [PMID: 39531618 DOI: 10.1513/annalsats.202404-382oc] [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: 04/16/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024] Open
Abstract
Rationale: Although exposure to air pollution is a known risk factor for adverse pulmonary outcomes, its impact in individuals with idiopathic pulmonary fibrosis (IPF) is less well understood. Objectives: To investigate the effects of long-term exposure to air pollution on disease severity and progression in patients with IPF and to determine whether genomic factors, such as MUC5B promoter polymorphism or telomere length, modify these associations. Methods: We performed analyses at enrollment and after 1 year of follow-up in the IPF-PRO (Idiopathic Pulmonary Fibrosis Prospective Outcomes) Registry, a prospective observational registry that enrolled individuals with IPF at 46 U.S. sites from June 2014 to October 2018. Five-year average pollution exposures (particulate matter ≤2.5 μm in aerodynamic diameter [PM2.5], nitrogen dioxide, ozone) before the enrollment date were estimated at participants' residential addresses with validated national spatiotemporal models. Multivariable regression models estimated associations between pollution exposure and physiologic measurements (forced vital capacity [FVC], diffusing capacity of the lung for carbon monoxide, supplemental oxygen use at rest) and quality-of-life measurements (St. George's Respiratory Questionnaire, EuroQoL, Cough and Sputum Assessment Questionnaire) at enrollment. Cox proportional hazards models estimated associations between pollutants and a composite outcome of death, lung transplant, or >10% absolute decline in FVC percent predicted in the year after enrollment. Models were adjusted for individual-level and spatial confounders, including proxies for disease onset. Gene-environment interactions with MUC5B and telomere length were assessed. Results: Of 835 participants, 94% were non-Hispanic White individuals, 76% were male, and the mean (standard deviation) age was 70 (7.7) years. In fully adjusted analyses, higher PM2.5 exposure was associated with worse quality of life per St. George's Respiratory Questionnaire activity score (3.48 [95% confidence interval (CI), 0.64, 6.32] per 2 μg/m3 PM2.5) and EuroQoL scores (-0.04 [95% CI, -0.06, -0.01] per 2 μg/m3 PM2.5), as well as lower FVC percent predicted and lower diffusing capacity of the lung for carbon monoxide percent predicted at enrollment. Each 3 parts per billion difference in O3 exposure was associated with a 1.57% (95% CI, 0.15, 2.98) higher FVC percent predicted at enrollment, although this effect was attenuated in multipollutant models. There was no association between nitrogen dioxide and enrollment measures or between pollution exposure and 1-year outcomes and no evidence for gene-environment interactions. Conclusions: In the IPF-PRO Registry, long-term exposure to PM2.5 was associated with worse quality of life and lung function at enrollment, but not with short-term disease progression or mortality. There was no evidence of effect modification by interaction of genomic factors with pollution. The reason for the unexpected relationship between O3 exposure and higher FVC is unclear. Clinical trial registered with www.clinicaltrials.gov (NCT01915511).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Peide Li
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut; and
| | - Amy L Olson
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut; and
| | - Thomas B Leonard
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut; and
| | - Jamie L Todd
- Duke Clinical Research Institute, Durham, North Carolina
- Duke University Medical Center, Durham, North Carolina
| | - Megan L Neely
- Duke Clinical Research Institute, Durham, North Carolina
- Duke University Medical Center, Durham, North Carolina
| | - Laurie D Snyder
- Duke Clinical Research Institute, Durham, North Carolina
- Duke University Medical Center, Durham, North Carolina
| | | |
Collapse
|
7
|
Cheng PP, He XL, Jia ZH, Hu SH, Feng X, Jiang YH, Li Q, Zhao LQ, Cui XL, Ye SY, Liang LM, Song LJ, Wang M, Yu F, Xiong L, Xiang F, Wang X, Ma WL, Ye H. Midkine, a novel MCP-1 activator mediated PM2.5-aggravated experimental pulmonary fibrosis. ENVIRONMENT INTERNATIONAL 2025; 197:109354. [PMID: 40049042 DOI: 10.1016/j.envint.2025.109354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/21/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025]
Abstract
Exposure to fine particulate matter (PM2.5) is associated with increased morbidity and mortality among patients with idiopathic pulmonary fibrosis (IPF). Pathological alterations in IPF typically originate in the subpleural regions of the lungs. However, it was unclear how PM2.5 affected subpleural pulmonary fibrosis. In this study, atmospheric PM2.5 and carbon blacks were utilized as representative particulate matter to investigate these effects. Mouse models and cell models were made to investigate macrophage chemotaxis changes under PM2.5 exposure in vivo and in vitro. The findings indicated that PM2.5 promoted macrophage aggregation in the subpleural region of lung and aggravated bleomycin-induced pulmonary fibrosis in mice. At the same time, we uncovered for the first time that PM2.5 exposure led to an upregulation of midkine, which subsequently enhanced the production of monocyte chemotactic protein-1 (MCP-1) through the cell surface receptor Syndecan 4 (SDC4) in pleural mesothelial cells (PMCs), thereby, inducing macrophage aggregation in subpleural region of lung. Furthermore, our results indicated that PM2.5 and bleomycin facilitated macrophage M1 polarization and the production of profibrotic inflammatory factors, culminating in fibrotic alterations in PMCs, lung fibroblasts, and alveolar epithelial cells. Finally, we demonstrated that inhibition of midkine ameliorated lung function and mitigated pulmonary fibrosis in vivo. In conclusion, our findings elucidated that midkine acted as a novel MCP-1 activator, mediating PM2.5-aggravated experimental pulmonary fibrosis, and suggested that the midkine/SDC4/MCP-1 signal should be a new therapeutic target for the treatment of PM2.5-related IPF.
Collapse
Affiliation(s)
- Pei-Pei Cheng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xin-Liang He
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, China
| | - Zi-Heng Jia
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shi-He Hu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao Feng
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ye-Han Jiang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qian Li
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li-Qin Zhao
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiao-Lin Cui
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shu-Yi Ye
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Li-Mei Liang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lin-Jie Song
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, China
| | - Meng Wang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fan Yu
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, China
| | - Liang Xiong
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, China
| | - Fei Xiang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, China
| | - Xiaorong Wang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, China
| | - Wan-Li Ma
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, China.
| | - Hong Ye
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Respiratory Diseases of National Health Commission of China, Wuhan, China.
| |
Collapse
|
8
|
Wang Y, Tian F, Qian ZM, Feng J, Wang X, McMillin SE, Howard SW, Lin H. Air pollution, metabolic signatures, and the risk of idiopathic pulmonary fibrosis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 964:178409. [PMID: 39837121 DOI: 10.1016/j.scitotenv.2025.178409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/16/2024] [Accepted: 01/04/2025] [Indexed: 01/23/2025]
Abstract
Air pollution has been associated with a higher incidence of idiopathic pulmonary fibrosis (IPF), yet this metabolic mechanism remains unclear. 185,865 participants were included in the UK Biobank. We estimated air pollution exposure using the bilinear interpolation approach, including fine particle matter with diameter < 2.5 μm (PM2.5), particle matter with diameter < 10 μm (PM10), nitrogen dioxide (NO2), and nitrogen oxides (NOx). We identified metabolites and established the metabolic signature with air pollutants using an elastic net regularized regression. Cox proportional hazards models combined with generalized propensity score (GPS) were conducted to evaluate the relationships between metabolic signatures and incident IPF, and mediation analysis was performed to evaluate potential mediators. During a median follow-up of 12.3 years, 1239 IPF cases were ascertained. We identified multi-metabolite profiles comprising 87 metabolites for PM2.5, 65 metabolites for PM10, 71 metabolites for NO2, and 76 metabolites for NOx. Metabolic signatures were associated with incident IPF, with HRs of 1.20 (95 % CI: 1.13, 1.27), 1.09 (95 % CI: 1.03, 1.15), 1.23 (95 % CI: 1.16, 1.31), and 1.24 (95 % CI: 1.17, 1.31) per standard deviation (SD) increase in metabolic profiles associated with PM2.5, PM10, NO2, and NOx, respectively. Furthermore, metabolic signatures of PM2.5, PM10, NO2 and NOx significantly mediated 5.71 %, 3.98 %, 4.21 %, and 4.58 % of air pollution on IPF. Long-term air pollution was associated with a higher risk of IPF, with metabolites potentially playing a mediating role. The findings emphasize the significance of improving metabolic status for the prevention of IPF.
Collapse
Affiliation(s)
- Yuhua Wang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Fei Tian
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Zhengmin Min Qian
- Department of Epidemiology and Biostatistics, College for Public Health & Social Justice, Saint Louis University, 3545 Lafayette Avenue, Saint Louis, MO 63104, USA
| | - Jin Feng
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Xiaojie Wang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Stephen Edward McMillin
- School of Social Work, Saint Louis University, Tegeler Hall, 3550 Lindell Boulevard, Saint Louis, MO 63103, USA
| | - Steven W Howard
- Department of Health Services Administration, School of Health Professions, University of Alabama at Birmingham, 1716 9th Avenue South, Birmingham, AL 35233, USA
| | - Hualiang Lin
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, China.
| |
Collapse
|
9
|
Xu M, Chen J, Gao L, Cai S, Dong H. Microplastic exposure induces HSP90α secretion and aggravates asthmatic airway remodeling via PI3K-Akt-mTOR pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 291:117828. [PMID: 39923560 DOI: 10.1016/j.ecoenv.2025.117828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/21/2025] [Accepted: 01/29/2025] [Indexed: 02/11/2025]
Abstract
Microplastics pollution has raised a considerable awareness due to their extensive distribution in the environment. It has potential side effects on human health. Microplastics can enter the human respiratory system, then deposit in the lung, destroying the structure of the bronchus and alveoli, and causing pulmonary inflammation, mucus production, and airway hyperresponsiveness, leading to the aggravation of asthma. Nevertheless, the underlying mechanism remains elusive. There are several cytokines involved in the inflammatory response of asthma. Heat shock protein 90α(HSP90α) is one of cytokines involving in inflammation which is a member of the HSPs family. The aim of this study is to explore the mechanism by which microplastics influence the secretion of HSP90α and the progression of asthma. Initially, we found that microplastics were destroyed airway epithelial barrier, resulting in inherent dysfunction in the secretion of HSP90α. Then, microplastics were proved to activate PI3K-Akt-mTOR pathway by prompting airway epithelial cells secrete HSP90α and proliferation of airway smooth muscle cells(ASMCs), leading to airway narrowing and hypersensitivity. 1G6-D7 is a monoclonal antibody to HSP90, which can reverse the pulmonary inflammation infiltration, mucus production, and airway hyperresponsiveness(AHR). Overall, these finding suggested that microplastics elicited inflammation via the PI3K-Akt-mTOR signaling pathway and stimulated the proliferation of ASMCs. Hence, the present study unveils a novel mechanism responsible for microplastic-induced inflammation and airway hyperreactivity, establishing a basis for further research and risk evaluations of microplastics.
Collapse
Affiliation(s)
- Mingming Xu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jiyuan Chen
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Lin Gao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shaoxi Cai
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Hangming Dong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
10
|
Xing M, Ma Y, Cui F, Li D, Wang J, Tang L, Zheng L, Yang J, Tian Y. Reply. Arthritis Rheumatol 2025; 77:116-117. [PMID: 39187473 DOI: 10.1002/art.42980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Affiliation(s)
- Meiqi Xing
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yudiyang Ma
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feipeng Cui
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dankang Li
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianing Wang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linxi Tang
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zheng
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Yang
- The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital and Hubei Key Laboratory of Ischemic Cardiovascular Disease and Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Yaohua Tian
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Jo YS, Song JW. Air Pollution and Interstitial Lung Disease. Tuberc Respir Dis (Seoul) 2025; 88:45-55. [PMID: 39542009 PMCID: PMC11704737 DOI: 10.4046/trd.2024.0116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/11/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024] Open
Abstract
This review article explores the multifaceted relationship between air pollution and interstitial lung diseases (ILDs), particularly focusing on idiopathic pulmonary fibrosis, the most severe form of fibrotic ILD. Air pollutants are mainly composed of particulate matter, ozone (O3), nitrogen dioxide (NO2), carbon monoxide (CO), and sulfur dioxide (SO2). They are recognized as risk factors for several respiratory diseases. However, their specific effects on ILDs and related mechanisms have not been thoroughly studied yet. Emerging evidence suggests that air pollutants may contribute to the development and acute exacerbation of ILDs. Longitudinal studies have indicated that air pollution can adversely affect the prognosis of disease by decreasing lung function and increasing mortality. Lots of in vitro, in vivo , and epidemiologic studies have proposed possible mechanisms linking ILDs to air pollution, including inflammation and oxidative stress induced by exposure to air pollutants, which may induce mitochondrial dysfunction, promote cellular senescence, and disrupt normal epithelial repair processes. Despite these findings, effective interventions to mitigate effects of air pollution on ILD are not well established yet. This review emphasizes the urgent need to address air pollution as a key environmental risk factor for ILDs and calls for further studies to clarify its effects and develop preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Yong Suk Jo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin Woo Song
- Department of Pulmonology and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
12
|
Moodley Y. Contemporary Concise Review 2023: Interstitial lung disease. Respirology 2024; 29:1095-1100. [PMID: 39438044 DOI: 10.1111/resp.14848] [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: 09/26/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
In this review, we have discussed several important developments in 2023 in Interstitial Lung Disease (ILD). The association of pollution with genetic predispositions increased the risk of Idiopathic Pulmonary Fibrosis (IPF). An interesting comorbidity of malnutrition was not adequately recognized in ILD. Novel genes have been identified in IPF involving predominantly short telomere length and surfactant protein production leading to alveolar epithelial cell dysfunction. Genetics also predicted progression in IPF. Crosstalk between vascular endothelial cells and fibroblasts in IPF mediated by bone morphogenic protein signalling may be important for remodelling of the lung. A novel modality for monitoring of disease included the 4-min gait speed. New treatment modalities include inhaled pirfenidone, efzofitimod, for sarcoidosis, and earlier use of immunosuppression in connective tissue disease-ILD.
Collapse
Affiliation(s)
- Yuben Moodley
- School of Medicine, University of Western Australia, Crawley, Western Australia, Australia
- Institute for Respiratory Health, Nedlands, Western Australia, Australia
- Department of Respiratory Medicine, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
- Centre for Research Excellence in Pulmonary Fibrosis, Australia
| |
Collapse
|
13
|
Fang M, He B, Xiao J. Smoking and Idiopathic Pulmonary Fibrosis: A Meta-analysis. Nicotine Tob Res 2024; 26:1599-1606. [PMID: 38666790 DOI: 10.1093/ntr/ntae100] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 03/18/2024] [Accepted: 04/23/2024] [Indexed: 12/06/2024]
Abstract
INTRODUCTION In this study, we aimed to systematically explore the relationship between smoking and idiopathic pulmonary fibrosis (IPF). AIMS AND METHODS The PubMed, Web of Science, and Embase databases were searched to systematically identify eligible studies. The Newcastle‒Ottawa Quality Assessment Scale (NOS) was used to evaluate the quality of the selected studies. The pooled odds ratio (OR) and survival hazard ratio (HR) were calculated with a random effects model using Stata 16.0 software. RESULTS Thirty studies were enrolled. All of the included studies were considered to have intermediate or high quality. Nine studies were suitable for meta-analysis of ORs, and 21 studies were suitable for meta-analysis of survival HR. The pooled analysis revealed a significant difference in the risk of IPF between the smoking group and the never-smoking group (OR 1.71, 95% CI: 1.27 to 2.30, p < .001), indicating that smoking is a risk factor for IPF. When analyzing pooled survival HRs, never smoking was compared to former smoking or current smoking. Former smoking was shown to be a poor prognostic factor for IPF (HR 1.43, 95% CI: 1.18 to 1.74, p < .001), but current smoking was not a significant factor. CONCLUSIONS Our results indicated that smoking is a risk factor for IPF patients. IMPLICATIONS In this study, we mainly concluded that smoking is a risk factor for IPF and that former smoking is a poor prognostic factor for IPF. To the best of our knowledge, this is the first meta-analysis report focusing on the association between smoking per se and IPF. Through our current study, we hope to further raise awareness of the relationship between smoking and IPF.
Collapse
Affiliation(s)
- Min Fang
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China
- School of Pharmacy, Changsha Medical University, Changsha, China
| | - Bixiu He
- Department of Geriatrics and Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Xiao
- Department of Geriatrics and Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
14
|
He YS, Ge M, Xu YQ, Gao ZX, He T, Zhang P, Tao SS, Wang P, Chen Z, Pan HF. Associations between blue space exposure and rheumatoid arthritis: The modifying effect of genetic susceptibility and air pollutants. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 287:117346. [PMID: 39541697 DOI: 10.1016/j.ecoenv.2024.117346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/16/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024]
Abstract
Studies on the interaction among genetic susceptibility, blue space exposure, and rheumatoid arthritis (RA) risk have been lacking. Therefore, we examined the association between blue space exposure and RA incidence and assess the modifying effect of genetic susceptibility and air pollutants. Form the UK Biobank, 322,783 participants without RA were enrolled in this study. The association between blue space exposure and RA incidence was estimated using a cox proportional hazards model. The combined effect of blue space and genetic factors on the risk of RA was further evaluated. The polygenic risk score (PRS) for RA was calculated to represent individual genetic risk, and the potential modification effect of air pollution on the relationship between blue space, PRS, and RA were explored. During a median follow-up of 12.4 years, 3659 RA cases were identified. A 10 % increase in blue space300 m was associated with a 22.6 % reduction in RA incidence (HR=0.774, 95 % CI: 0.670, 0.895), exhibiting a consistent downward trend in the exposure-response curve. A high PRS was an independent risk factor for RA (HR=1.393, 95 % CI: 1.347, 1.439). The associations between blue space exposure, PRS, and the risk of RA were dose-dependent, with the lowest risk observed among those with high levels of blue space and lower PRS (HRbluespace300m=0.501, 95 % CI: 0.431, 0.583; HRbluespace1000m=0.476, 95 % CI: 0.408, 0.555). Interaction analysis indicated that increased concentrations of air pollutants strengthened the relationship between PRS and RA. Blue space exposure mitigated the risk of RA development, particularly in individuals with low genetic risk.
Collapse
Affiliation(s)
- Yi-Sheng He
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity-mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Man Ge
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity-mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Yi-Qing Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity-mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Zhao-Xing Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity-mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Tian He
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity-mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Peng Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity-mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Sha-Sha Tao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity-mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Peng Wang
- Department of Health Promotion and Behavioral Sciences, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Zhu Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China.
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity-mediated Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Center for Big Data and Population Health of IHM, Anhui Medical University, Hefei, Anhui 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China.
| |
Collapse
|
15
|
Ning Z, Ma Y, He S, Li G, Xu Y, Wang Z, Zhang Y, Ma E, Ma C, Wu J. High altitude air pollution and respiratory disease: Evaluating compounded exposure events and interactions. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117046. [PMID: 39276646 DOI: 10.1016/j.ecoenv.2024.117046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Today, air pollution remains a significant issue, particularly in high-altitude areas where its impact on respiratory disease remains incompletely explored. This study aims to investigate the association between various air pollutants and outpatient visits for respiratory disease in such regions, specifically focussing on Xining from 2016 to 2021. By analysing over 570,000 outpatient visits using a time-stratified case-crossover design and conditional logistic regression, we assessed the independent effects of pollutants like PM2.5, PM10, SO2, NO2, and CO, as well as their interactions. The evaluation of interactions employed measures such as relative excess odds due to interaction (REOI), attributable proportion due to interaction (AP), and synergy index (S). We also conducted a stratified analysis to identify potentially vulnerable populations. Our findings indicated that exposure to PM2.5, PM10, SO2, NO2, and CO significantly increased outpatient visits for respiratory disease, with odds ratios (ORs) of 2.40 % (95 % CI: 2.05 %, 2.74 %), 1.07 % (0.98 %, 1.16 %), 3.86 % (3.23 %, 4.49 %), 4.45 % (4.14 %, 4.77 %), and 6.37 % (5.70 %, 7.04 %), respectively. However, exposure to O3 did not show a significant association. We found significant interactions among PM2.5, SO2, NO2, and CO, where combined exposure further exacerbated the risk of respiratory diseases. For example, in the combination of PM2.5 and SO2, the REOI, AP, and S were 0.07 (95 % CI: 0.06, 0.09), 0.07 (0.06, 0.07), and 1.07 (1.05, 1.09), respectively. Additionally, elderly individuals and females were more sensitive to these pollutants, but no statistically significant interaction effects were observed between different age and gender groups. In conclusion, our study highlights the strong link between air pollution and respiratory disease in high-altitude areas, with combined pollutant exposure posing an even greater risk. It underscores the need for enhanced air quality monitoring and public awareness campaigns, particularly to protect vulnerable populations like the elderly and females.
Collapse
Affiliation(s)
- Zhenxu Ning
- Department of Public Health, Qinghai University Medical College, Xining, Qinghai 810016, China
| | - Yanjun Ma
- Qinghai Institute of Health Sciences, Xining, Qinghai 810016, China.
| | - Shuzhen He
- Xining Centre for Disease Control and Prevention, Xining, Qinghai 810000, China.
| | - Genshan Li
- Department of Public Health, Qinghai University Medical College, Xining, Qinghai 810016, China
| | - Yueshun Xu
- Qinghai Meteorological Bureau, Xining, Qinghai 810000, China
| | - Zhanqing Wang
- Datong County Center for Disease Control and Prevention, Xining, Qinghai 810100, China
| | - Yunxia Zhang
- The First People's Hospital of Xining, Xining, Qinghai 810000, China
| | - Enzhou Ma
- Qinghai Meteorological Bureau, Xining, Qinghai 810000, China
| | - Chunguang Ma
- Xining Centre for Disease Control and Prevention, Xining, Qinghai 810000, China
| | - Jing Wu
- Xining Centre for Disease Control and Prevention, Xining, Qinghai 810000, China
| |
Collapse
|
16
|
Meng P, Pan C, Qin X, Cai Q, Zhao Y, Wei W, Cheng S, Yang X, Cheng B, Liu L, He D, Shi S, Chu X, Zhang N, Jia Y, Wen Y, Liu H, Zhang F. A genome-wide gene-environmental interaction study identified novel loci for the relationship between ambient air pollution exposure and depression, anxiety. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117121. [PMID: 39357380 DOI: 10.1016/j.ecoenv.2024.117121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 09/11/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Genetic factors and environmental exposures, including air pollution, contribute to the risk of depression and anxiety. While the association between air pollution and depression and anxiety has been established in the UK Biobank, there has been limited research exploring this relationship from a genetic perspective. METHODS Based on individual genotypic and phenotypic data from a cohort of 104,385 participants in the UK Biobank, a polygenic risk score for depression and anxiety was constructed to explore the joint effects of nitric oxide (NO), nitrogen dioxide (NO2), particulate matter (PM) with a diameter of ⩽2.5 μm (PM2.5) and 2.5-10 μm (PMcoarse) with depression and anxiety by linear and logistic regression models. Subsequently, a genome-wide gene-environmental interaction study (GWEIS) was performed using PLINK 2.0 to identify the genes interacting with air pollution for depression and anxiety. RESULTS A substantial risk of depression and anxiety development was detected in participants exposed to the high air pollution concomitantly with high genetic risk. GWEIS identified 166, 23, 18, and 164 significant candidate loci interacting with NO, NO2, PM2.5, and PMcoarse for Patient Health Questionnaire-9 (PHQ-9) score, and detected 44, 10, 10, and 114 candidate loci associated with NO, NO2, PM2.5, and PMcoarse for General Anxiety Disorder (GAD-7) score, respectively. And some significant genes overlapped among four air pollutants, like TSN (rs184699498, PNO2 = 3.47 × 10-9; rs139212326, PPM2.5 = 1.51 × 10-8) and HSP90AB7P(rs150987455, PNO2 = 1.63 × 10-11; rs150987455, PPM2.5 = 7.64 × 10-11), which were common genes affecting PHQ-9 score for both NO2 and PM2.5. CONCLUSION Our study identified the joint effects of air pollution with genetic susceptibility on the risk of depression and anxiety, and provided several novel candidate genes for the interaction, contributing to an understanding of the genetic architecture of depression and anxiety.
Collapse
Affiliation(s)
- Peilin Meng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Chuyu Pan
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyue Qin
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Qingqing Cai
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yijing Zhao
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Wenming Wei
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Shiqiang Cheng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xuena Yang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Bolun Cheng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Li Liu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Dan He
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Sirong Shi
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoge Chu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Na Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yumeng Jia
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yan Wen
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Huan Liu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China.
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
17
|
Wang J, Li K, Hao D, Li X, Zhu Y, Yu H, Chen H. Pulmonary fibrosis: pathogenesis and therapeutic strategies. MedComm (Beijing) 2024; 5:e744. [PMID: 39314887 PMCID: PMC11417429 DOI: 10.1002/mco2.744] [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: 02/18/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/25/2024] Open
Abstract
Pulmonary fibrosis (PF) is a chronic and progressive lung disease characterized by extensive alterations of cellular fate and function and excessive accumulation of extracellular matrix, leading to lung tissue scarring and impaired respiratory function. Although our understanding of its pathogenesis has increased, effective treatments remain scarce, and fibrotic progression is a major cause of mortality. Recent research has identified various etiological factors, including genetic predispositions, environmental exposures, and lifestyle factors, which contribute to the onset and progression of PF. Nonetheless, the precise mechanisms by which these factors interact to drive fibrosis are not yet fully elucidated. This review thoroughly examines the diverse etiological factors, cellular and molecular mechanisms, and key signaling pathways involved in PF, such as TGF-β, WNT/β-catenin, and PI3K/Akt/mTOR. It also discusses current therapeutic strategies, including antifibrotic agents like pirfenidone and nintedanib, and explores emerging treatments targeting fibrosis and cellular senescence. Emphasizing the need for omni-target approaches to overcome the limitations of current therapies, this review integrates recent findings to enhance our understanding of PF and contribute to the development of more effective prevention and management strategies, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Jianhai Wang
- Department of Respiratory MedicineHaihe HospitalTianjin UniversityTianjinChina
- Department of TuberculosisHaihe HospitalTianjin UniversityTianjinChina
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese MedicineTianjin Institute of Respiratory DiseasesTianjinChina
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe HospitalTianjin UniversityTianjinChina
| | - Kuan Li
- Department of Respiratory MedicineHaihe HospitalTianjin UniversityTianjinChina
- Department of TuberculosisHaihe HospitalTianjin UniversityTianjinChina
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe HospitalTianjin UniversityTianjinChina
| | - De Hao
- Department of Respiratory MedicineHaihe HospitalTianjin UniversityTianjinChina
| | - Xue Li
- Department of Respiratory MedicineHaihe HospitalTianjin UniversityTianjinChina
- Department of TuberculosisHaihe HospitalTianjin UniversityTianjinChina
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe HospitalTianjin UniversityTianjinChina
| | - Yu Zhu
- Department of Clinical LaboratoryNankai University Affiliated Third Central HospitalTianjinChina
- Department of Clinical LaboratoryThe Third Central Hospital of TianjinTianjin Key Laboratory of Extracorporeal Life Support for Critical DiseasesArtificial Cell Engineering Technology Research Center of TianjinTianjin Institute of Hepatobiliary DiseaseTianjinChina
| | - Hongzhi Yu
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe HospitalTianjin UniversityTianjinChina
| | - Huaiyong Chen
- Department of Respiratory MedicineHaihe HospitalTianjin UniversityTianjinChina
- Department of TuberculosisHaihe HospitalTianjin UniversityTianjinChina
- Key Research Laboratory for Infectious Disease Prevention for State Administration of Traditional Chinese MedicineTianjin Institute of Respiratory DiseasesTianjinChina
- Tianjin Key Laboratory of Lung Regenerative Medicine, Haihe HospitalTianjin UniversityTianjinChina
| |
Collapse
|
18
|
Huang P, Wang X, Cao Y, Yang J, Yao R, Liang L, Cheng G, Yang L. Research progress on the use of Salvia miltiorrhiza Bunge extracts in the treatment of pulmonary diseases. Biomed Pharmacother 2024; 179:117282. [PMID: 39146764 DOI: 10.1016/j.biopha.2024.117282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/01/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024] Open
Abstract
Salvia miltiorrhiza Bunge extracts, known for their diverse biological activities, often have remarkable efficacy in treating pulmonary diseases overlooked due to their specific cardiovascular actions. With the recent outbreak of COVID-19, research into pulmonary-related diseases has garnered significant attention. Salvia miltiorrhiza Bunge extracts can be broadly categorized into lipophilic and hydrophilic components; however, a comprehensive summary of their mechanisms in treating pulmonary diseases is lacking. Therefore, this review aims to systematically summarize the therapeutic mechanisms of 10 major Salvia miltiorrhiza Bunge extracts in treating pulmonary fibrosis, lung cancer, acute lung injury, and chronic obstructive pulmonary disease, with the goal of identifying promising options for efficacious therapies.
Collapse
Affiliation(s)
- Peifeng Huang
- School of Integrative medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xuezhen Wang
- School of Integrative medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yingyi Cao
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Jiaming Yang
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rongmei Yao
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Leiqin Liang
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518000, China.
| | - Long Yang
- School of Integrative medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
19
|
Liu T, Ning J, Fan X, Wei H, Shi G, Fu QB. Identification of immune patterns in idiopathic pulmonary fibrosis patients driven by PLA2G7-positive macrophages using an integrated machine learning survival framework. Sci Rep 2024; 14:22369. [PMID: 39333367 PMCID: PMC11437001 DOI: 10.1038/s41598-024-73625-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: 02/13/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024] Open
Abstract
Patients with advanced idiopathic pulmonary fibrosis (IPF), a complex and incurable lung disease with an elusive pathology, are nearly exclusive candidates for lung transplantation. Improved identification of patient subtypes can enhance early diagnosis and intervention, ultimately leading to better prognostic outcomes for patients. The goal of this study is to identify new immune patterns and biomarkers in patients. Immune subtypes in IPF patients were identified using single-sample gene set enrichment analysis, and immune subtype-related genes were explored using the weighted correlation network analysis algorithm. A machine learning integration framework was used to establish the optimal prognostic model, known as the immune-related risk score (IRS). Single-cell sequencing was conducted to investigate the major role of macrophage-derived PLA2G7 in the immune microenvironment. We assessed the stability of celecoxib in targeting PLA2G7 through molecular docking and surface plasmon resonance. IPF patients present two distinct immune subtypes, one characterized by immune activation and inflammation, and the other by immune suppression. IRS can predict the immune status and prognosis of IPF patients. Furthermore, multi-cohort analysis and single-cell sequencing analysis demonstrated the diagnostic and prognostic value of PLA2G7 derived from macrophages and its role in shaping the inflammatory immune microenvironment in IPF patients. Celecoxib could effectively and stably bind with PLA2G7. PLA2G7, as identified through IRS, demonstrates marked stability in diagnosing and predicting the prognosis of IPF patients as well as predicting their immune status. It can serve as a novel biomarker for IPF patients.
Collapse
Affiliation(s)
- Tianxi Liu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jingyuan Ning
- Department of Immunology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Xiaoqing Fan
- Institute of Microbiological Testing and Inspection, Tianjin Centre for Disease Control and Prevention, Tianjin , People's Republic of China
| | - Huan Wei
- Department of Neurology, The Affiliated Yan'an Hospital of Kunming Medical University, Kunming, People's Republic of China
| | - Guangsen Shi
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, People's Republic of China.
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Qingshan Bill Fu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong, People's Republic of China.
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
20
|
Liu J, Song J, Li Y, Gao D, Ma Q, Song X, Jiang J, Zhang Y, Wang R, Dong Z, Chen L, Qin Y, Yuan W, Guo T, Song Z, Dong Y, Zou Z, Ma J. Geneenvironment interaction between phthalate exposure and pubertal genetic polymorphisms on blood pressure variability in children: Exploring the moderating effects of lifestyle behaviours. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116966. [PMID: 39216218 DOI: 10.1016/j.ecoenv.2024.116966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Phthalates (PAEs) are synthetic compounds extensively employed in consumer products. Blood pressure (BP) in children can vary, the degree of visit-to-visit BP variability (VVV) is at least partially independent of BP. The interactions between PAEs exposure, pubertal-related genetic susceptibility and lifestyles on childhood VVV are not investigated. This study utilized data from a cohort collected from Oct 2017-2020 in Xiamen, China. Seven urine PAE metabolites were measured. The long-term VVV was characterized employing the standard deviation (SD) and average real variability. We constructed a genetic risk score (GRS) of pubertal-related genes and healthy lifestyle scores. Exposed to high levels of mono-2-ethyl-5-hydroxyhexyl phthalate (MEHHP) (OR=1.43, 95 %CI=1.07, 1.92) and mono-2-ethyl-5-oxohexyl phthalate (OR=1.36, 95 % CI=1.01, 1.83) was related to increased SBP-SD, and the OR for high SBP-SD related to high GRS was 1.38 (95 % CI=1.02, 1.85). Compared to participants who had low GRS and low MEHHP exposure, participants exhibiting high GRS and MEHHP levels were more likely to experience high SBP-SD (OR=2.00, P<0.05). Individuals exhibiting low GRS, low MEHHP levels, and adhering to healthy lifestyles were associated with the least probability of experiencing high SBP-SD (OR=0.31, P<0.05). Increased PAEs exposure could elevate childhood systolic VVV, and exacerbated the adverse impact of pubertal-related genetic susceptibility on the high VVV of SBP; however, healthy lifestyles might alleviate these adverse effects. Promoting healthy lifestyles and reducing PAEs exposure for preventing elevated BP variability among children is important, especially for individuals with greater genetic susceptibility to early pubertal onset. ENVIRONMENTAL IMPLICATION: Blood pressure (BP) in children can vary, as a noninvasive, inexpensive and applicable method, the extent of visit-to-visit variability (VVV) is at least partially independent of BP. The interactions between phthalates (PAEs) exposure, variants of puberty-related genes and lifestyles on VVV are not investigated. Increased childhood systolic VVV might be associated with PAEs exposure, with the associations more pronounced combined with pubertal genetic susceptibility. Yet, healthy habits could partly eliminate such adverse effects. Our study underscores the importance of advocating for healthy lifestyles and reducing exposure to PAEs, especially among individuals with high genetic susceptibility to early puberty onset.
Collapse
Affiliation(s)
- Jieyu Liu
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Jieyun Song
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Yanhui Li
- School of Nursing, Peking University, Beijing, China
| | - Di Gao
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Qi Ma
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Xinli Song
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Jianuo Jiang
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Yi Zhang
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Ruolin Wang
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Ziqi Dong
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Li Chen
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Yang Qin
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Wen Yuan
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Tongjun Guo
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Zhiying Song
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| | - Yanhui Dong
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China.
| | - Zhiyong Zou
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China.
| | - Jun Ma
- Institute of Child and Adolescent Health, School of Public Health, Peking University, National Health Commission Key Laboratory of Reproductive Health, Beijing, China
| |
Collapse
|
21
|
Baddour NA, Paulin LM, Gassett AJ, Woo H, Hoffman EA, Newell JD, Woodruff PG, Pirozzi CS, Barjaktarevic I, Barr RG, O’Neal W, Han MK, Martinez FJ, Peters SP, Hastie AT, Hansel NN, Ortega VE, Kaufman JD, Sack CS. Air Pollution Exposure and Interstitial Lung Features in SPIROMICS Participants with Chronic Obstructive Pulmonary Disease. Ann Am Thorac Soc 2024; 21:1251-1260. [PMID: 38568439 PMCID: PMC11376362 DOI: 10.1513/annalsats.202308-741oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 04/02/2024] [Indexed: 08/31/2024] Open
Abstract
Rationale: It is unknown whether air pollution is associated with radiographic features of interstitial lung disease in individuals with chronic obstructive pulmonary disease (COPD). Objectives: To determine whether air pollution increases the prevalence of interstitial lung abnormalities (ILA) or percent high-attenuation areas (HAA) on computed tomography (CT) in individuals with a heavy smoking history and COPD. Methods: We performed a cross-sectional study of SPIROMICS (Subpopulations and Intermediate Outcome Measures in COPD Study), focused on current or former smokers with COPD. Ten-year exposure to particulate matter ⩽2.5 μm in aerodynamic diameter (PM2.5), nitrogen oxides (NOx), nitrogen dioxide (NO2), and ozone before enrollment CT (completed between 2010 and 2015) were estimated with validated spatiotemporal models at residential addresses. We applied adjusted multivariable modified Poisson regression and linear regression to investigate associations between pollution exposure and relative risk (RR) of ILA or increased percent HAA (between -600 and -250 Hounsfield units), respectively. We assessed for effect modification by MUC5B-promoter polymorphism (variant allele carriers GT or TT vs. GG at rs3705950), smoking status, sex, and percent emphysema. Results: Among 1,272 participants with COPD assessed for HAA, 424 were current smokers, and 249 were carriers of the variant MUC5B allele. A total of 519 participants were assessed for ILA. We found no association between pollution exposure and ILA or HAA. Associations between pollutant exposures and risk of ILA were modified by the presence of MUC5B polymorphism (P value interaction term for NOx = 0.04 and PM2.5 = 0.05) and smoking status (P value interaction term for NOx = 0.05; NO2 = 0.01; and ozone = 0.05). With higher exposure to NOx and PM2.5, MUC5B variant carriers had an increased risk of ILA (RR per 26 ppb NOx, 2.41; 95% confidence interval [CI], 0.97-6.0; and RR per 4 μg ⋅ m-3 PM2.5, 1.43; 95% CI, 0.93-2.2, respectively). With higher exposure to NO2, former smokers had an increased risk of ILA (RR per 10 ppb, 1.64; 95% CI, 1.0-2.7). Conclusions: Exposure to ambient air pollution was not associated with interstitial features on CT in this population of heavy smokers with COPD. MUC5B modified the association between pollution and ILA, suggesting that gene-environment interactions may influence prevalence of interstitial lung features in COPD.
Collapse
Affiliation(s)
| | - Laura M. Paulin
- Department of Medicine, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire
| | | | - Han Woo
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Eric A. Hoffman
- Department of Radiology, University of Iowa, Iowa City, Iowa
| | - John D. Newell
- Department of Radiology, University of Washington, Seattle, Washington
- Department of Radiology, University of Iowa, Iowa City, Iowa
| | - Prescott G. Woodruff
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California
| | - Cheryl S. Pirozzi
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, Department of Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Igor Barjaktarevic
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - R. Graham Barr
- Department of Medicine, College of Physicians and Surgeons, Columbia University Medical Center, New York, New York
| | - Wanda O’Neal
- Marsico Lung Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Meilan K. Han
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Fernando J. Martinez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medical College, Cornell University, New York, New York
| | - Stephen P. Peters
- Section of Pulmonary, Critical Care, Allergy and Immunologic Diseases, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina; and
| | - Annette T. Hastie
- Section of Pulmonary, Critical Care, Allergy and Immunologic Diseases, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina; and
| | - Nadia N. Hansel
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Victor E. Ortega
- Division of Respiratory Diseases, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona
| | - Joel D. Kaufman
- Department of Medicine
- Department of Environmental and Occupational Health Sciences, and
| | - Coralynn S. Sack
- Department of Medicine
- Department of Environmental and Occupational Health Sciences, and
| |
Collapse
|
22
|
Tang L, Li D, Wang J, Su B, Tian Y. Ambient air pollution, genetic risk and telomere length in UK biobank. JOURNAL OF EXPOSURE SCIENCE & ENVIRONMENTAL EPIDEMIOLOGY 2024; 34:845-852. [PMID: 37550565 DOI: 10.1038/s41370-023-00587-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND Telomere length (TL) is a biomarker of genomic aging. The evidence on the association between TL and air pollution was inconsistent. Besides, the modification effect of genetic susceptibility on the air pollution-TL association remains unknown. OBJECTIVE We aimed to evaluate the association of ambient air pollution with TL and further assess the modification effect of genetic susceptibility. METHODS 433,535 participants with complete data of TL and air pollutants in UK Biobank were included. Annual average exposure of NO2, NOx, PM10 and PM2.5 was estimated by applying land use regression models. Genetic risk score (GRS) was constructed using reported telomere-related SNPs. Leukocyte TL was measured by quantitative polymerase chain reaction (qPCR). Multivariable linear regression models were employed to conduct associational analyses. RESULTS Categorical exposure models and RCS models both indicated U-shaped (for NO2 and NOx) and L-shaped (for PM10 and PM2.5) correlations between air pollution and TL. In comparison to the lowest quartile, the 2nd and 3rd quartile of NO2 (q2: -1.3% [-2.1%, -0.4%]; q3: -1.2% [-2.0%, -0.3%], NOx (q2: -1.3% [-2.1%, -0.5%]; q3: -1.4% [-2.2%, -0.5%]), PM2.5 (q2: -0.8% [-1.7%, 0.0%]; q3: -1.3% [-2.2%, -0.5%]), and the third quartile of PM10 (q3: -1.1% [-1.9%, -0.2%]) were inversely associated with TL. The highest quartile of NO2 was positively correlated with TL (q4: 1.0% [0.0%, 2.0%]), whereas the negative correlation between the highest quartile of other pollutants and TL was also attenuated and no longer significant. In the genetic analyses, synergistic interactions were observed between the 4th quartile of three air pollutants (NO2, NOx, and PM2.5) and genetic risk. IMPACT STATEMENT Our study for the first time revealed a non-linear trend for the association between air pollution and telomere length. The genetic analyses suggested synergistic interactions between air pollution and genetic risk on the air pollution-TL association. These findings may shed new light on air pollution's health effects, offer suggestions for identifying at-risk individuals, and provide hints regarding further investigation into gene-environment interactions.
Collapse
Affiliation(s)
- Linxi Tang
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, 430030, Wuhan, China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, 430030, Wuhan, China
| | - Dankang Li
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, 430030, Wuhan, China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, 430030, Wuhan, China
| | - Jianing Wang
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, 430030, Wuhan, China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, 430030, Wuhan, China
| | - Binbin Su
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences/Peking Union Medical College, No.31, Beijige-3, Dongcheng District, 100730, Beijing, China.
| | - Yaohua Tian
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, 430030, Wuhan, China.
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, 430030, Wuhan, China.
| |
Collapse
|
23
|
Marimoutou M, Patel V, Kim JH, Schaible N, Alvarez J, Hughes J, Obermok M, Rodríguez CI, Kallarakal T, Suki B, Amin K, Krishnan R, Behrsing HP. The Fibrotic Phenotype of Human Precision-Cut Lung Slices Is Maintained after Cryopreservation. TOXICS 2024; 12:637. [PMID: 39330565 PMCID: PMC11436228 DOI: 10.3390/toxics12090637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024]
Abstract
Human precision-cut lung slices (hPCLS) prepared from fibrotic lungs recapitulate the pathophysiological hallmarks of fibrosis. These hallmark features can also be induced by treating non-fibrotic hPCLS with a fibrotic cocktail (FC). As a result, the fibrotic and fibrosis-induced hPCLS are rapidly emerging as preferred models for disease modeling and drug discovery. However, current hPCLS models are limited by tissue viability in culture, as they are usually only viable for one week after harvesting. Here, we demonstrate that the fibrotic hPCLS can be cryopreserved, stored for months, and then thawed on demand without loss of hPCLS viability or protein content for 14 days post-thawing. Cryopreservation also preserves the pro-fibrotic potential of non-fibrotic hPCLS. Specifically, when we treated the thawed non-fibrotic hPCLS with an FC, we observed significant pro-fibrotic cytokine secretion and elevated tissue stiffness. These pro-fibrotic changes were inhibited by the small-molecule tyrosine kinase inhibitor, Nintedanib. Taken together, our work indicates that a feasible solution to prolong the pre-clinical utility of fibrotic and fibrosis-induced hPCLS is cryopreservation. We anticipate that cryopreserved hPCLS will serve as an advantageous predictive model for the evaluation of pro-fibrotic pathways during acute and chronic toxicity testing.
Collapse
Affiliation(s)
- Méry Marimoutou
- Institute for In Vitro Sciences, Inc., Gaithersburg, MD 20878, USA
| | - Vivek Patel
- Institute for In Vitro Sciences, Inc., Gaithersburg, MD 20878, USA
| | - Jae Hun Kim
- Mechanobiologix, LLC, Newton, MA 02464, USA
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Niccole Schaible
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Jose Alvarez
- Institute for In Vitro Sciences, Inc., Gaithersburg, MD 20878, USA
| | - Joseph Hughes
- Institute for In Vitro Sciences, Inc., Gaithersburg, MD 20878, USA
| | - McKenzie Obermok
- Institute for In Vitro Sciences, Inc., Gaithersburg, MD 20878, USA
| | | | | | - Béla Suki
- Mechanobiologix, LLC, Newton, MA 02464, USA
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Khalid Amin
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ramaswamy Krishnan
- Mechanobiologix, LLC, Newton, MA 02464, USA
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | | |
Collapse
|
24
|
Adegunsoye A, Kropski JA, Behr J, Blackwell TS, Corte TJ, Cottin V, Glanville AR, Glassberg MK, Griese M, Hunninghake GM, Johannson KA, Keane MP, Kim JS, Kolb M, Maher TM, Oldham JM, Podolanczuk AJ, Rosas IO, Martinez FJ, Noth I, Schwartz DA. Genetics and Genomics of Pulmonary Fibrosis: Charting the Molecular Landscape and Shaping Precision Medicine. Am J Respir Crit Care Med 2024; 210:401-423. [PMID: 38573068 PMCID: PMC11351799 DOI: 10.1164/rccm.202401-0238so] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/04/2024] [Indexed: 04/05/2024] Open
Abstract
Recent genetic and genomic advancements have elucidated the complex etiology of idiopathic pulmonary fibrosis (IPF) and other progressive fibrotic interstitial lung diseases (ILDs), emphasizing the contribution of heritable factors. This state-of-the-art review synthesizes evidence on significant genetic contributors to pulmonary fibrosis (PF), including rare genetic variants and common SNPs. The MUC5B promoter variant is unusual, a common SNP that markedly elevates the risk of early and established PF. We address the utility of genetic variation in enhancing understanding of disease pathogenesis and clinical phenotypes, improving disease definitions, and informing prognosis and treatment response. Critical research gaps are highlighted, particularly the underrepresentation of non-European ancestries in PF genetic studies and the exploration of PF phenotypes beyond usual interstitial pneumonia/IPF. We discuss the role of telomere length, often critically short in PF, and its link to progression and mortality, underscoring the genetic complexity involving telomere biology genes (TERT, TERC) and others like SFTPC and MUC5B. In addition, we address the potential of gene-by-environment interactions to modulate disease manifestation, advocating for precision medicine in PF. Insights from gene expression profiling studies and multiomic analyses highlight the promise for understanding disease pathogenesis and offer new approaches to clinical care, therapeutic drug development, and biomarker discovery. Finally, we discuss the ethical, legal, and social implications of genomic research and therapies in PF, stressing the need for sound practices and informed clinical genetic discussions. Looking forward, we advocate for comprehensive genetic testing panels and polygenic risk scores to improve the management of PF and related ILDs across diverse populations.
Collapse
Affiliation(s)
- Ayodeji Adegunsoye
- Pulmonary/Critical Care, and
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois
| | - Jonathan A. Kropski
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Juergen Behr
- Department of Medicine V, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, member of the German Center for Lung Research (DZL), Munich, Germany
| | - Timothy S. Blackwell
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Tamera J. Corte
- Centre of Research Excellence in Pulmonary Fibrosis, Camperdown, New South Wales, Australia
- Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Vincent Cottin
- National Reference Center for Rare Pulmonary Diseases (OrphaLung), Louis Pradel Hospital, Hospices Civils de Lyon, ERN-LUNG (European Reference Network on Rare Respiratory Diseases), Lyon, France
- Claude Bernard University Lyon, Lyon, France
| | - Allan R. Glanville
- Lung Transplant Unit, St. Vincent’s Hospital Sydney, Sydney, New South Wales, Australia
| | - Marilyn K. Glassberg
- Department of Medicine, Loyola Chicago Stritch School of Medicine, Chicago, Illinois
| | - Matthias Griese
- Department of Pediatric Pneumology, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians-University, German Center for Lung Research, Munich, Germany
| | - Gary M. Hunninghake
- Harvard Medical School, Boston, Massachusetts
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | | | - Michael P. Keane
- Department of Respiratory Medicine, St. Vincent’s University Hospital and School of Medicine, University College Dublin, Dublin, Ireland
| | - John S. Kim
- Department of Medicine, School of Medicine, and
| | - Martin Kolb
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Toby M. Maher
- Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, California
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Justin M. Oldham
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | | | | | - Fernando J. Martinez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York; and
| | - Imre Noth
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - David A. Schwartz
- Department of Medicine, School of Medicine, University of Colorado, Aurora, Colorado
| |
Collapse
|
25
|
Zhang S, Li X, Zhang L, Zhang Z, Li X, Xing Y, Wenger JC, Long X, Bao Z, Qi X, Han Y, Prévôt ASH, Cao J, Chen Y. Disease types and pathogenic mechanisms induced by PM 2.5 in five human systems: An analysis using omics and human disease databases. ENVIRONMENT INTERNATIONAL 2024; 190:108863. [PMID: 38959566 DOI: 10.1016/j.envint.2024.108863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/05/2024]
Abstract
Atmospheric fine particulate matter (PM2.5) can harm various systems in the human body. Due to limitations in the current understanding of epidemiology and toxicology, the disease types and pathogenic mechanisms induced by PM2.5 in various human systems remain unclear. In this study, the disease types induced by PM2.5 in the respiratory, circulatory, endocrine, and female and male urogenital systems have been investigated and the pathogenic mechanisms identified at molecular level. The results reveal that PM2.5 is highly likely to induce pulmonary emphysema, reperfusion injury, malignant thyroid neoplasm, ovarian endometriosis, and nephritis in each of the above systems respectively. The most important co-existing gene, cellular component, biological process, molecular function, and pathway in the five systems targeted by PM2.5 are Fos proto-oncogene (FOS), extracellular matrix, urogenital system development, extracellular matrix structural constituent conferring tensile strength, and ferroptosis respectively. Differentially expressed genes that are significantly and uniquely targeted by PM2.5 in each system are BTG2 (respiratory), BIRC5 (circulatory), NFE2L2 (endocrine), TBK1 (female urogenital) and STAT1 (male urogenital). Important disease-related cellular components, biological processes, and molecular functions are specifically induced by PM2.5. For example, response to wounding, blood vessel morphogenesis, body morphogenesis, negative regulation of response to endoplasmic reticulum stress, and response to type I interferon are the top uniquely existing biological processes in each system respectively. PM2.5 mainly acts on key disease-related pathways such as the PD-L1 expression and PD-1 checkpoint pathway in cancer (respiratory), cell cycle (circulatory), apoptosis (endocrine), antigen processing and presentation (female urogenital), and neuroactive ligand-receptor interaction (male urogenital). This study provides a novel analysis strategy for elucidating PM2.5-related disease types and is an important supplement to epidemiological investigation. It clarifies the risks of PM2.5 exposure, elucidates the pathogenic mechanisms, and provides scientific support for promoting the precise prevention and treatment of PM2.5-related diseases.
Collapse
Affiliation(s)
- Shumin Zhang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Xiaomeng Li
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China; Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China; Department of Laboratory Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Liru Zhang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Zhengliang Zhang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China; School of Public Health, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Xuan Li
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China; School of Public Health, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Yan Xing
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - John C Wenger
- School of Chemistry and Environmental Research Institute, University College Cork, Cork, Ireland
| | - Xin Long
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Zhier Bao
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Xin Qi
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Yan Han
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - André S H Prévôt
- Laboratory of Atmospheric Chemistry, Paul Scherrer Institut, Villigen, PSI 5232, Switzerland
| | - Junji Cao
- Institute of Atmospheric Physics, Chinese Academy of Sciences, Beijing 100029, China
| | - Yang Chen
- Research Center for Atmospheric Environment, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China.
| |
Collapse
|
26
|
Moos PJ, Cheminant JR, Cowman S, Noll J, Wang Q, Musci T, Venosa A. Spatial and phenotypic heterogeneity of resident and monocyte-derived macrophages during inflammatory exacerbations leading to pulmonary fibrosis. Front Immunol 2024; 15:1425466. [PMID: 39100672 PMCID: PMC11294112 DOI: 10.3389/fimmu.2024.1425466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/28/2024] [Indexed: 08/06/2024] Open
Abstract
Introduction Genetic mutations in critical nodes of pulmonary epithelial function are linked to the pathogenesis of pulmonary fibrosis (PF) and other interstitial lung diseases. The slow progression of these pathologies is often intermitted and accelerated by acute exacerbations, complex non-resolving cycles of inflammation and parenchymal damage, resulting in lung function decline and death. Excess monocyte mobilization during the initial phase of an acute exacerbation, and their long-term persistence in the lung, is linked to poor disease outcome. Methods The present work leverages a clinical idiopathic PF dataset and a murine model of acute inflammatory exacerbations triggered by mutation in the alveolar type-2 cell-restricted Surfactant Protein-C [SP-C] gene to spatially and phenotypically define monocyte/macrophage changes in the fibrosing lung. Results SP-C mutation triggered heterogeneous CD68+ macrophage activation, with highly active peri-injured cells relative to those sampled from fully remodeled and healthy regions. Ingenuity pathway analysis of sorted CD11b-SigF+CD11c+ alveolar macrophages defined asynchronous activation of extracellular matrix re-organization, cellular mobilization, and Apolipoprotein E (Apoe) signaling in the fibrosing lung. Cell-cell communication analysis of single cell sequencing datasets predicted pro-fibrogenic signaling (fibronectin/Fn1, osteopontin/Spp1, and Tgfb1) emanating from Trem2/TREM2 + interstitial macrophages. These cells also produced a distinct lipid signature from alveolar macrophages and monocytes, characterized by Apoe expression. Mono- and di-allelic genetic deletion of ApoE in SP-C mutant mice had limited impact on inflammation and mortality up to 42 day after injury. Discussion Together, these results provide a detailed spatio-temporal picture of resident, interstitial, and monocyte-derived macrophages during SP-C induced inflammatory exacerbations and end-stage clinical PF, and propose ApoE as a biomarker to identify activated macrophages involved in tissue remodeling.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Alessandro Venosa
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, UT, United States
| |
Collapse
|
27
|
Zhang Q, Meng X, Luo H, Yu K, Li A, Zhou L, Chen R, Kan H. Air pollutants, genetic susceptibility, and incident schizophrenia in later life: A prospective study in the UK Biobank. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 934:173009. [PMID: 38734111 DOI: 10.1016/j.scitotenv.2024.173009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/10/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
OBJECTIVE Air pollution has been linked to multiple psychiatric disorders, but little is known on its long-term association with schizophrenia. The interaction between air pollution and genetic susceptibility on incident schizophrenia has never been reported. We aimed to explore the associations between long-term air pollution exposure and late-onset schizophrenia and evaluate whether genetic susceptibility could modify the association. METHODS This population-based prospective cohort study included 437,802 middle-aged and elderly individuals free of schizophrenia at baseline in the UK Biobank. Land use regression models were applied in the estimation of the annual average concentrations of nitrogen dioxide (NO2), nitrogen oxides (NOx), fine particulate matter (PM2.5), and inhalable particulate matter (PM10) at residence. The associations between air pollutants and schizophrenia were evaluated by using Cox proportional hazard models. A polygenic risk score of schizophrenia was constructed for exploring potential interaction of air pollutants with genetic susceptibility. RESULTS An interquartile range increase in PM2.5, PM10, NO2, and NOx was associated with the hazard ratios (HR) for incident schizophrenia at 1.19, 1.16, 1.22, and 1.09, respectively. The exposure-response curves for the association of air pollution with incident schizophrenia were approximately linear. There are additive interactions of air pollution score (APS), PM10, NO2, and NOx with genetic risk. Specifically, compared with participants with low genetic susceptibility and low APS, the HR was 3.23 for individuals with high genetic risk and high APS, among which 0.49 excess risk could be attributed to the additive interaction, accounting for 15 % of the schizophrenia risk. CONCLUSION This large-scale, prospective cohort study conveys the first-hand evidence that long-term air pollution exposure could elevate schizophrenia incidence in later life, especially for individuals with higher genetic risks. The findings highlight the importance of improving air quality for preventing the late-onset schizophrenia in an aging era, especially among those with high genetic risks.
Collapse
Affiliation(s)
- Qingli Zhang
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, China; Ministry of Education - Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Meng
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, China
| | - Huihuan Luo
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, China
| | - Kexin Yu
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, China
| | - Anni Li
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, China
| | - Lu Zhou
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, China
| | - Renjie Chen
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, China; School of Public Health, University of South China, Hengyang, Hunan, China; School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan, China..
| | - Haidong Kan
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai, China; Children's Hospital of Fudan University, National Center for Children's Health, Shanghai, China.
| |
Collapse
|
28
|
Liu Z, Peng Z, Lin H, Zhou K, Liang L, Cao J, Huang Z, Mei J. Identifying potential drug targets for idiopathic pulmonary fibrosis: a mendelian randomization study based on the druggable genes. Respir Res 2024; 25:217. [PMID: 38783236 PMCID: PMC11118997 DOI: 10.1186/s12931-024-02848-5] [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: 09/26/2023] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a chronic fibrotic interstitial lung disease characterized by progressive dyspnea and decreased lung function, yet its exact etiology remains unclear. It is of great significance to discover new drug targets for IPF. METHODS We obtained the cis-expression quantitative trait locus (cis-eQTL) of druggable genes from eQTLGen Consortium as exposure and the genome wide association study (GWAS) of IPF from the International IPF Genetics Consortium as outcomes to simulate the effects of drugs on IPF by employing mendelian randomization analysis. Then colocalization analysis was performed to calculate the probability of both cis-eQTL of druggable genes and IPF sharing a causal variant. For further validation, we conducted protein quantitative trait locus (pQTL) analysis to reaffirm our findings. RESULTS The expression of 45 druggable genes was significantly associated with IPF susceptibility at FDR < 0.05. The expression of 23 and 15 druggable genes was significantly associated with decreased forced vital capacity (FVC) and diffusing capacity of the lungs for carbon monoxide (DLco) in IPF patients, respectively. IPF susceptibility and two significant genes (IL-7 and ABCB2) were likely to share a causal variant. The results of the pQTL analysis demonstrated that high levels of IL-7 in plasma are associated with a reduced risk of IPF (OR = 0.67, 95%CI: 0.47-0.97). CONCLUSION IL-7 stands out as the most promising potential drug target to mitigate the risk of IPF. Our study not only sheds light on potential drug targets but also provides a direction for future drug development in IPF.
Collapse
Affiliation(s)
- Zetao Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Zhiyu Peng
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Huahang Lin
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Ke Zhou
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Linchuan Liang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Jie Cao
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Zhaokang Huang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China
| | - Jiandong Mei
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Western China Collaborative Innovation Center for Early Diagnosis and Multidisciplinary Therapy of Lung Cancer, Sichuan University, Chengdu, China.
| |
Collapse
|
29
|
Cui F, Tang L, Li D, Ma Y, Wang J, Xie J, Su B, Tian Y, Zheng X. Early-life exposure to tobacco, genetic susceptibility, and accelerated biological aging in adulthood. SCIENCE ADVANCES 2024; 10:eadl3747. [PMID: 38701212 PMCID: PMC11068008 DOI: 10.1126/sciadv.adl3747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 04/03/2024] [Indexed: 05/05/2024]
Abstract
Early-life tobacco exposure serves as a non-negligible risk factor for aging-related diseases. To understand the underlying mechanisms, we explored the associations of early-life tobacco exposure with accelerated biological aging and further assessed the joint effects of tobacco exposure and genetic susceptibility. Compared with those without in utero exposure, participants with in utero tobacco exposure had an increase in Klemera-Doubal biological age (KDM-BA) and PhenoAge acceleration of 0.26 and 0.49 years, respectively, but a decrease in telomere length of 5.34% among 276,259 participants. We also found significant dose-response associations between the age of smoking initiation and accelerated biological aging. Furthermore, the joint effects revealed that high-polygenic risk score participants with in utero exposure and smoking initiation in childhood had the highest accelerated biological aging. There were interactions between early-life tobacco exposure and age, sex, deprivation, and diet on KDM-BA and PhenoAge acceleration. These findings highlight the importance of reducing early-life tobacco exposure to improve healthy aging.
Collapse
Affiliation(s)
- Feipeng Cui
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030, Hubei, PR China
| | - Linxi Tang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030, Hubei, PR China
| | - Dankang Li
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030, Hubei, PR China
| | - Yudiyang Ma
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030, Hubei, PR China
| | - Jianing Wang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030, Hubei, PR China
| | - Junqing Xie
- Center for Statistics in Medicine, NDORMS, University of Oxford, The Botnar Research Centre, Oxford, UK
| | - Binbin Su
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences/Peking Union Medical College, No. 31, Beijige-3, Dongcheng District, Beijing 100730, PR China
| | - Yaohua Tian
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030, Hubei, PR China
| | - Xiaoying Zheng
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences/Peking Union Medical College, No. 31, Beijige-3, Dongcheng District, Beijing 100730, PR China
| |
Collapse
|
30
|
Sesé L, Annesi-Maesano I. Lung cancer and idiopathic pulmonary fibrosis: environmental exposures are the key. Eur Respir J 2024; 63:2400760. [PMID: 38816038 DOI: 10.1183/13993003.00760-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 05/03/2024] [Indexed: 06/01/2024]
Affiliation(s)
- Lucile Sesé
- Department of Physiology and Functional Explorations, AP-HP, Hôpital Avicenne, INSERM UMR 1272 "Hypoxia and the Lung", Université Sorbonne Paris Nord, Bobigny, France
- Department of Pneumology, Constitutive Reference Center for Rare Lung Diseases, AP-HP, Hôpital Avicenne, Bobigny, France
| | - Isabella Annesi-Maesano
- Desbrest Institute of Epidemiology and Public Health, Univ Montpellier, INSERM, Montpellier, France
- Division of Respiratory Medicine, Allergology, and of Thoracic Oncology, University Hospital of Montpellier, Montpellier, France
| |
Collapse
|
31
|
Goobie GC, Saha PK, Carlsten C, Gibson KF, Johannson KA, Kass DJ, Ryerson CJ, Zhang Y, Robinson AL, Presto AA, Nouraie SM. Ambient Ultrafine Particulate Matter and Clinical Outcomes in Fibrotic Interstitial Lung Disease. Am J Respir Crit Care Med 2024; 209:1082-1090. [PMID: 38019094 PMCID: PMC11092946 DOI: 10.1164/rccm.202307-1275oc] [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: 07/26/2023] [Accepted: 11/28/2023] [Indexed: 11/30/2023] Open
Abstract
Rationale: Particulate matter with an aerodynamic diameter ⩽2.5 μm is associated with adverse outcomes in fibrotic interstitial lung disease (fILD), but the impact of ultrafine particulates (UFPs; aerodynamic diameter ⩽100 nm) remains unknown. Objective: To evaluate UFP associations with clinical outcomes in fILD. Methods: We conducted a multicenter, prospective cohort study enrolling patients with fILD from the University of Pittsburgh Dorothy P. and Richard P. Simmons Center and the Pulmonary Fibrosis Foundation Patient Registry (PFF-PR). Using a national-scale UFP model, we linked exposures using three approaches in the Simmons cohort (residential address geocoordinates, ZIP code centroid geocoordinates, and ZIP code average) and two in the PFF-PR for which only five-digit ZIP code was available (ZIP code centroid and ZIP code average). We tested UFP associations with transplantation-free survival using multivariable Cox proportional-hazards models, baseline percentage predicted FVC and DlCO using multivariable linear regressions, and decline in FVC and DlCO using linear mixed models adjusting for age, sex, smoking, race, socioeconomic status, site, particulate matter with an aerodynamic diameter ⩽2.5, and nitrogen dioxide. Measurements and Main Results: Annual mean outdoor UFP concentrations for 2017 were estimated for 1,416 Simmons and 1,919 PFF-PR patients. Increased UFP concentration was associated with transplantation-free survival in fully adjusted Simmons residential address models (hazard ratio, 1.08 per 1,000 particles/cm3 [95% confidence interval, 1.01-1.15]; P = 0.02) but not PFF-PR models, which used less precise linkage approaches. Higher UFP exposure was associated with lower baseline FVC and more rapid FVC decline in the Simmons registry. Conclusions: Increased UFP exposure was associated with transplantation-free survival and lung function in the cohort with precise residential location linkage. This work highlights the need for more robust regulatory networks to study the health effects of UFPs nationwide.
Collapse
Affiliation(s)
- Gillian C. Goobie
- Centre for Heart Lung Innovation and
- St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Provat K. Saha
- Center for Atmospheric Particle Studies and
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
- Department of Civil Engineering, Bangladesh University of Engineering and Technology, Dhaka, Bangladesh
| | - Christopher Carlsten
- Centre for Heart Lung Innovation and
- St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Air Pollution Exposure Laboratory, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada; and
| | - Kevin F. Gibson
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kerri A. Johannson
- Division of Respiratory Medicine, Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Daniel J. Kass
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christopher J. Ryerson
- Centre for Heart Lung Innovation and
- St. Paul’s Hospital, Vancouver, British Columbia, Canada
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yingze Zhang
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Allen L. Robinson
- Center for Atmospheric Particle Studies and
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Albert A. Presto
- Center for Atmospheric Particle Studies and
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - S. Mehdi Nouraie
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
32
|
Li SR, Kang NN, Wang RR, Li MD, Chen LH, Zhou P, Xu DX, Zhao H, Fu L. ALKBH5 SUMOylation-mediated FBXW7 m6A modification regulates alveolar cells senescence during 1-nitropyrene-induced pulmonary fibrosis. JOURNAL OF HAZARDOUS MATERIALS 2024; 468:133704. [PMID: 38364577 DOI: 10.1016/j.jhazmat.2024.133704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/21/2024] [Accepted: 01/31/2024] [Indexed: 02/18/2024]
Abstract
Our previous study revealed that 1-nitropyrene (1-NP) exposure evoked pulmonary fibrosis in mice. However, the exact mechanism remained elusive. We found that 1-NP induced telomere damage and cellular senescence in mice lungs, and two alveolar epithelial cells lines. 1-NP downregulated telomere repeat binding factor 2 (TRF2), and upregulated FBXW7. Mechanistically, 1-NP-caused TRF2 ubiquitination and proteasomal degradation depended on E3 ubiquitin ligase activity of FBXW7. Moreover, 1-NP upregulated FBXW7 m6A modification via an ALKBH5-YTHDF1-dependent manner. Further analysis suggested 1-NP promoted ALKBH5 SUMOylation and subsequent proteasomal degradation. Additionally, 1-NP evoked mitochondrial reactive oxygen species (mtROS) overproduction. Mito-TEMPO, a mitochondrial-targeted antioxidant, mitigated 1-NP-caused mtROS overproduction, ALKBH5 SUMOylation, FBXW7 m6A modification, TRF2 degradation, cellular senescence, and pulmonary fibrosis. Taken together, mtROS-initiated ALKBH5 SUMOylation and subsequent FBXW7 m6A modification is indispensable for TRF2 degradation and cellular senescence in alveolar epithelial cells during 1-NP-induced pulmonary fibrosis. Our study provides target intervention measures towards 1-NP-evoked pulmonary fibrosis.
Collapse
Affiliation(s)
- Se-Ruo Li
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Ning-Ning Kang
- Department of Thoracic Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Rong-Rong Wang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Meng-Die Li
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Li-Hong Chen
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Peng Zhou
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei, Anhui 230032, China
| | - Hui Zhao
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China.
| | - Lin Fu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Institute of Respiratory Diseases, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China; Department of Toxicology, Anhui Medical University, Hefei, Anhui 230032, China.
| |
Collapse
|
33
|
Tan T, Tang L, Guo X, Li T, Tian Y, Ouyang Z. Associations of residential greenness with bone mineral density and osteoporosis: the modifying effect of genetic susceptibility. Ann Rheum Dis 2024; 83:669-676. [PMID: 38443139 DOI: 10.1136/ard-2023-224941] [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: 08/30/2023] [Accepted: 01/03/2024] [Indexed: 03/07/2024]
Abstract
OBJECTIVES To investigate the associations of residential greenness with bone mineral density and incident osteoporosis, and further evaluate the potential modifying effect of genetic susceptibility. METHODS We used the Normalised Difference Vegetation Index (NDVI) at various buffer distances, including 300 m (NDVI300m), 500 m (NDVI500m), 1000 m (NDVI1000m) and 1500 m (NDVI1500m), to serve as indicators of greenness. We fitted linear regression, logistic regression and Cox proportional hazard models to assess the associations of residential greenness with estimated bone mineral density (eBMD), prevalent osteoporosis and incident osteoporosis, respectively. With the Polygenic Risk Score (PRS) for osteoporosis, we further assessed the joint effects of genetic risk and greenness on the risk of osteoporosis. We conducted causal mediation analyses to explore potential mediators. RESULTS Each IQR increase in NDVI300m was associated with 0.0007 (95% CI 0.0002 to 0.0013) increase in eBMD, 6% lower risk of prevalent osteoporosis (OR 0.94; 95% CI 0.92 to 0.97) and 5% lower risk of incident osteoporosis (HR 0.95; 95% CI 0.93 to 0.98). The joint effects of greenness and PRS on the risk of osteoporosis displayed a clear dose-response pattern. Compared with individuals exposed to low NDVI levels and high genetic risk, those exposed to high NDVI levels and low genetic risk had a 56% (95% CI 51% to 61%) lower risk of osteoporosis. The primary mediators in the association between greenness and incident osteoporosis were identified as PM2.5 and NO2. CONCLUSIONS Residential greenness was associated with higher bone mineral density and decreased risk of incident osteoporosis.
Collapse
Affiliation(s)
- Tingting Tan
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Linxi Tang
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoning Guo
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tao Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Osteopathy Laboratory of Surgical,The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yaohua Tian
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengxiao Ouyang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Osteopathy Laboratory of Surgical,The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
34
|
Yao XI, Tong X, Shen C, Song Y, Sun S, Chen K, Shen H. Green space, genetic susceptibility, and risk of osteoporosis:a cohort study from the UK Biobank. CHEMOSPHERE 2024; 353:141632. [PMID: 38442776 DOI: 10.1016/j.chemosphere.2024.141632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
OBJECTIVE This study aimed to investigate the effect of residential exposure to green space on the incident osteoporosis and further explore the modification effect of genetic susceptibility. METHODS Participants from the UK Biobank were followed from 2006 to 2010 (baseline) to December 31st, 2022. Using land use coverage, we evaluated exposure to residential surrounding green space, natural environment, and domestic gardens. We used the Cox regression to examine the association between the residential environment and incident osteoporosis. The interactive effects between polygenic risk score (PRS) of osteoporosis and residential environments on incident osteoporosis were investigated. RESULTS This study included 292,662 participants. Over a median follow-up period of 13.65 years, we documented 9177 incidents of osteoporosis. Per interquartile (IQR) increase in greenness and natural environment at a 300 m buffer was associated with a 4% lower risk of incident osteoporosis [HR = 0.96 (95% CI: 0.93, 0.99)] and [HR = 0.96 (95% CI: 0.93, 0.98)], respectively. We did not identify any interactive effects between genetic risk and residential environment on incident osteoporosis. CONCLUSIONS This study found that public greenness and natural environments could reduce the risk of incident osteoporosis regardless of genetic predisposition. Developing sustainable and publicly accessible natural environments might benefit populations' bone health.
Collapse
Affiliation(s)
- Xiaoxin I Yao
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, PR China; Department of Clinical Research, The Eighth Affiliated Hospital, Sun Yat-sen University, PR China
| | - Xinning Tong
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, PR China
| | - Chen Shen
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK; National Institute for Health Research Health Protection Research Unit in Chemical and Radiation Threats and Hazards, Imperial College London, UK
| | - Yichang Song
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, PR China
| | - Shengzhi Sun
- School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China.
| | - Keng Chen
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, PR China.
| | - Huiyong Shen
- Department of Orthopaedics, The Eighth Affiliated Hospital, Sun Yat-Sen University, PR China; Department of Clinical Research, The Eighth Affiliated Hospital, Sun Yat-sen University, PR China.
| |
Collapse
|
35
|
Tang L, Cui F, Ma Y, Li D, Wang J, Liu R, Tian Y. Residential greenness and incident idiopathic pulmonary fibrosis: A prospective study. ENVIRONMENTAL RESEARCH 2024; 245:117984. [PMID: 38154569 DOI: 10.1016/j.envres.2023.117984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/28/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND The impact of residential greenness on incident idiopathic pulmonary fibrosis (IPF) is unknown. We aimed to assess the association between residential greenness and incident IPF, identify underlying pathways, and further evaluate the effect among different genetic subgroups. METHODS 469,348 participants in the UK Biobank were included and followed until December 2020. Normalized difference vegetation index (NDVI) within 300-, 500-, 1000-, and 1500-m buffers (NDVI300m, NDVI500m, NDVI1000m, and NDVI1500m) were employed as indicators of greenness. The polygenic risk score (PRS) was constructed based on 13 independent SNPs. Cox models were fitted to assess the association of residential greenness with incident IPF. Casual mediation analyses were applied to evaluate potential mediators. FINDINGS After a median follow-up of 11.85 years, 1574 IPF cases were identified. We found residential greenness inversely associated with incident IPF. The HRs (95%CIs) for each interquartile increase of NDVI300m, NDVI500m, NDVI1000m, NDVI1500m were 0.93 (0.87, 0.99), 0.92 (0.86, 0.98), 0.89 (0.83, 0.95), and 0.89 (0.83, 0.95), respectively. The association was stronger among individuals with intermediate or high genetic risk. In mediation analyses, the main mediators identified were PM2.5 and NO2, with proportion mediated estimated to be 31.92% and 40.61% respectively for NDVI300m. INTERPRETATION Residential greenness was associated with reduced risk of incident IPF.
Collapse
Affiliation(s)
- Linxi Tang
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China; Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China
| | - Feipeng Cui
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China; Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China
| | - Yudiyang Ma
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China; Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China
| | - Dankang Li
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China; Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China
| | - Jianing Wang
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China; Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China
| | - Run Liu
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China; Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China
| | - Yaohua Tian
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China; Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China.
| |
Collapse
|
36
|
Li C, Feng X, Li S, He X, Luo Z, Cheng X, Yao J, Xiao J, Wang X, Wen D, Liu D, Li Y, Zhou H, Ma L, Lin T, Cai X, Lin Y, Guo L, Yang M. Tetrahedral DNA loaded siCCR2 restrains M1 macrophage polarization to ameliorate pulmonary fibrosis in chemoradiation-induced murine model. Mol Ther 2024; 32:766-782. [PMID: 38273656 PMCID: PMC10928155 DOI: 10.1016/j.ymthe.2024.01.022] [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: 09/07/2023] [Revised: 12/05/2023] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic lethal disease in the absence of demonstrated efficacy for preventing progression. Although macrophage-mediated alveolitis is determined to participate in myofibrotic transition during disease development, the paradigm of continuous macrophage polarization is still under-explored due to lack of proper animal models. Here, by integrating 2.5 U/kg intratracheal Bleomycin administration and 10 Gy thorax irradiation at day 7, we generated a murine model with continuous alveolitis-mediated fibrosis, which mimics most of the clinical features of our involved IPF patients. In combination with data from scRNA-seq of patients and a murine IPF model, a decisive role of CCL2/CCR2 axis in driving M1 macrophage polarization was revealed, and M1 macrophage was further confirmed to boost alveolitis in leading myofibroblast activation. Multiple sticky-end tetrahedral framework nucleic acids conjunct with quadruple ccr2-siRNA (FNA-siCCR2) was synthesized in targeting M1 macrophages. FNA-siCCR2 successfully blocked macrophage accumulation in pulmonary parenchyma of the IPF murine model, thus preventing myofibroblast activation and leading to the disease remitting. Overall, our studies lay the groundwork to develop a novel IPF murine model, reveal M1 macrophages as potential therapeutic targets, and establish new treatment strategy by using FNA-siCCR2, which are highly relevant to clinical scenarios and translational research in the field of IPF.
Collapse
Affiliation(s)
- Chen Li
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Xiaorong Feng
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Songhang Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xing He
- School of Clinical Medicine, Chengdu Medical College, Chengdu 610500, China
| | - Zeli Luo
- Department of Pulmonary and Critical Care Medicine, Wenjiang Hospital of Sichuan Provincial People's, Chengdu 611138, China
| | - Xia Cheng
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Jie Yao
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Jie Xiao
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Xiaofei Wang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dingke Wen
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Duanya Liu
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Yanfei Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Hong Zhou
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610056, China
| | - Lu Ma
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tongyu Lin
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; College of Biomedical Engineering, Sichuan University, Chengdu 610041, China.
| | - Lu Guo
- Department of Pulmonary and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China.
| | - Mu Yang
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610042, China.
| |
Collapse
|
37
|
Sesé L, Borie R, Kannengiesser C, Cottin V, Israel-Biet D, Crestani B, Cadranel J, Chenivesse C, Boubaya M, Valeyre D, Annesi-Maesano I, Nunes H. Impact of Air Pollution and MUC5B Genotype on Survival in Idiopathic Pulmonary Fibrosis. Ann Am Thorac Soc 2024; 21:519-523. [PMID: 38096447 DOI: 10.1513/annalsats.202305-495rl] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Affiliation(s)
- Lucile Sesé
- Hôpital Avicenne Bobigny, France
- Université Sorbonne Paris Nord Bobigny, France
| | - Raphaël Borie
- Hôpital Bichat Paris, France
- Université Paris Cité Paris, France
| | | | - Vincent Cottin
- Hôpital Louis Pradel Lyon, France
- Université Claude Bernard Lyon 1 Lyon, France
- Institut National de Recherche pour l'Agriculture et l'Environnement Paris, France
- OrphaLung, member of Respifil, ERN-LUNG Lyon, France
| | | | | | | | | | | | - Dominique Valeyre
- Hôpital Avicenne Bobigny, France
- Université Sorbonne Paris Nord Bobigny, France
| | | | - Hilario Nunes
- Université Sorbonne Paris Nord Bobigny, France
- Hôpital Avicenne Paris, France
| |
Collapse
|
38
|
Goobie GC. Neighborhood Disparities in Interstitial Lung Disease: How Do We Capture the Most Vulnerable? Ann Am Thorac Soc 2024; 21:377-380. [PMID: 38426828 PMCID: PMC10913766 DOI: 10.1513/annalsats.202311-959ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Affiliation(s)
- Gillian C Goobie
- Division of Respiratory Medicine, Department of Medicine, and Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; and Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
39
|
Zhang Y, Chen X, Fang D, Yan H, Wang D, Wang X, Li J, Zhai Y, Chu X, Wang D, Zhao H, Fang X. Adsorption Behavior of NO and NO 2 on Two-Dimensional As, Sb, and Bi Materials: First-Principles Insights. MATERIALS (BASEL, SWITZERLAND) 2024; 17:1024. [PMID: 38473496 DOI: 10.3390/ma17051024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 03/14/2024]
Abstract
To address the most significant environmental challenges, the quest for high-performance gas sensing materials is crucial. Among numerous two-dimensional materials, this study investigates the gas-sensitive capabilities of monolayer As, Sb, and Bi materials. To compare the gas detection abilities of these three materials, we employ first-principles calculations to comprehensively study the adsorption behavior of NO and NO2 gas molecules on the material surfaces. The results indicate that monolayer Bi material exhibits reasonable adsorption distances, substantial adsorption energies, and significant charge transfer for both NO and NO2 gases. Therefore, among the materials studied, it demonstrates the best gas detection capability. Furthermore, monolayer As and Sb materials exhibit remarkably high capacities for adsorbing NO and NO2 gas molecules, firmly interacting with the gas molecules. Gas adsorption induces changes in the material's work function, suggesting the potential application of these two materials as catalysts.
Collapse
Affiliation(s)
- Yuting Zhang
- State Key Laboratory of High Power Semiconductor Lasers, School of Physics, Changchun University of Science and Technology, 7089 Wei-Xing Road, Changchun 130022, China
| | - Xi Chen
- State Key Laboratory of High Power Semiconductor Lasers, School of Physics, Changchun University of Science and Technology, 7089 Wei-Xing Road, Changchun 130022, China
| | - Dan Fang
- State Key Laboratory of High Power Semiconductor Lasers, School of Physics, Changchun University of Science and Technology, 7089 Wei-Xing Road, Changchun 130022, China
| | - Hao Yan
- State Key Laboratory of High Power Semiconductor Lasers, School of Physics, Changchun University of Science and Technology, 7089 Wei-Xing Road, Changchun 130022, China
| | - Dengkui Wang
- State Key Laboratory of High Power Semiconductor Lasers, School of Physics, Changchun University of Science and Technology, 7089 Wei-Xing Road, Changchun 130022, China
| | - Xiaohua Wang
- State Key Laboratory of High Power Semiconductor Lasers, School of Physics, Changchun University of Science and Technology, 7089 Wei-Xing Road, Changchun 130022, China
| | - Jinhua Li
- State Key Laboratory of High Power Semiconductor Lasers, School of Physics, Changchun University of Science and Technology, 7089 Wei-Xing Road, Changchun 130022, China
| | - Yingjiao Zhai
- State Key Laboratory of High Power Semiconductor Lasers, School of Physics, Changchun University of Science and Technology, 7089 Wei-Xing Road, Changchun 130022, China
| | - Xueying Chu
- State Key Laboratory of High Power Semiconductor Lasers, School of Physics, Changchun University of Science and Technology, 7089 Wei-Xing Road, Changchun 130022, China
| | - Dongbo Wang
- Department of Opto-Electronic Information Science, School of Materials Science and Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Hongbin Zhao
- State Key Laboratory of High Power Semiconductor Lasers, School of Physics, Changchun University of Science and Technology, 7089 Wei-Xing Road, Changchun 130022, China
- State Key Laboratory of Advanced Materials for Smart Sensing, General Research Institute for Nonferrous Metals, Beijing 100088, China
| | - Xuan Fang
- State Key Laboratory of High Power Semiconductor Lasers, School of Physics, Changchun University of Science and Technology, 7089 Wei-Xing Road, Changchun 130022, China
| |
Collapse
|
40
|
Wang T, Duan W, Jia X, Huang X, Liu Y, Meng F, Ni C. Associations of combined phenotypic ageing and genetic risk with incidence of chronic respiratory diseases in the UK Biobank: a prospective cohort study. Eur Respir J 2024; 63:2301720. [PMID: 38061785 PMCID: PMC10882326 DOI: 10.1183/13993003.01720-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 11/29/2023] [Indexed: 02/24/2024]
Abstract
BACKGROUND Accelerated biological ageing has been associated with an increased risk of several chronic respiratory diseases. However, the associations between phenotypic age, a new biological age indicator based on clinical chemistry biomarkers, and common chronic respiratory diseases have not been evaluated. METHODS We analysed data from 308 592 participants at baseline in the UK Biobank. The phenotypic age was calculated from chronological age and nine clinical chemistry biomarkers, including albumin, alkaline phosphatase, creatinine, glucose, C-reactive protein, lymphocyte percent, mean cell volume, red cell distribution width and white blood cell count. Furthermore, phenotypic age acceleration (PhenoAgeAccel) was calculated by regressing phenotypic age on chronological age. The associations of PhenoAgeAccel with incident common chronic respiratory diseases and cross-sectional lung function were investigated. Moreover, we constructed polygenic risk scores and evaluated whether PhenoAgeAccel modified the effect of genetic susceptibility on chronic respiratory diseases and lung function. RESULTS The results showed significant associations of PhenoAgeAccel with increased risk of idiopathic pulmonary fibrosis (IPF) (hazard ratio (HR) 1.52, 95% CI 1.45-1.59), COPD (HR 1.54, 95% CI 1.51-1.57) and asthma (HR 1.18, 95% CI 1.15-1.20) per 5-year increase and decreased lung function. There was an additive interaction between PhenoAgeAccel and the genetic risk for IPF and COPD. Participants with high genetic risk and who were biologically older had the highest risk of incident IPF (HR 5.24, 95% CI 3.91-7.02), COPD (HR 2.99, 95% CI 2.66-3.36) and asthma (HR 2.07, 95% CI 1.86-2.31). Mediation analysis indicated that PhenoAgeAccel could mediate 10∼20% of the associations between smoking and chronic respiratory diseases, while ∼10% of the associations between particulate matter with aerodynamic diameter <2.5 µm and the disorders were mediated by PhenoAgeAccel. CONCLUSION PhenoAgeAccel was significantly associated with incident risk of common chronic respiratory diseases and decreased lung function and could serve as a novel clinical biomarker.
Collapse
Affiliation(s)
- Ting Wang
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang, China
| | - Weiwei Duan
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
- Joint first authors
- Joint first authors
| | - Xinying Jia
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xinmei Huang
- Department of Respiratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yi Liu
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
- Contributed equally to this article as lead authors and supervised the work
| | - Fanqing Meng
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Contributed equally to this article as lead authors and supervised the work
| | - Chunhui Ni
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang, China
- Contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
41
|
Samarelli AV, Tonelli R, Raineri G, Bruzzi G, Andrisani D, Gozzi F, Marchioni A, Costantini M, Fabbiani L, Genovese F, Pinetti D, Manicardi L, Castaniere I, Masciale V, Aramini B, Tabbì L, Rizzato S, Bettelli S, Manfredini S, Dominici M, Clini E, Cerri S. Proteomic profiling of formalin-fixed paraffine-embedded tissue reveals key proteins related to lung dysfunction in idiopathic pulmonary fibrosis. Front Oncol 2024; 13:1275346. [PMID: 38322285 PMCID: PMC10844556 DOI: 10.3389/fonc.2023.1275346] [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: 08/09/2023] [Accepted: 12/29/2023] [Indexed: 02/08/2024] Open
Abstract
Introduction Idiopathic pulmonary fibrosis (IPF) severely affects the lung leading to aberrant deposition of extracellular matrix and parenchymal stiffness with progressive functional derangement. The limited availability of fresh tissues represents one of the major limitations to study the molecular profiling of IPF lung tissue. The primary aim of this study was to explore the proteomic profiling yield of archived formalin-fixed paraffin-embedded (FFPE) specimens of IPF lung tissues. Methods We further determined the protein expression according to respiratory functional decline at the time of biopsy. The total proteins isolated from 11 FFPE samples of IPF patients compared to 3 FFPE samples from a non-fibrotic lung defined as controls, were subjected to label-free quantitative proteomic analysis by liquid chromatography-mass spectrometry (LC-MS/MS) and resulted in the detection of about 400 proteins. Results After the pairwise comparison between controls and IPF, functional enrichment analysis identified differentially expressed proteins that were involved in extracellular matrix signaling pathways, focal adhesion and transforming growth factor β (TGF-β) signaling pathways strongly associated with IPF onset and progression. Five proteins were significantly over- expressed in the lung of IPF patients with either advanced disease stage (Stage II) or impaired pulmonary function (FVC<75, DLCO<55) compared to controls; these were lymphocyte cytosolic protein 1 (LCP1), peroxiredoxin-2 (PRDX2), transgelin 2 (TAGLN2), lumican (LUM) and mimecan (OGN) that might play a key role in the fibrogenic processes. Discussion Our work showed that the analysis of FFPE samples was able to identify key proteins that might be crucial for the IPF pathogenesis. These proteins are correlated with lung carcinogenesis or involved in the immune landscape of lung cancer, thus making possible common mechanisms between lung carcinogenesis and fibrosis progression, two pathological conditions at risk for each other in the real life.
Collapse
Affiliation(s)
- Anna Valeria Samarelli
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Roberto Tonelli
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
- Respiratory Disease Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Giulia Raineri
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Giulia Bruzzi
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
- Respiratory Disease Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Dario Andrisani
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
- Respiratory Disease Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Filippo Gozzi
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
- Respiratory Disease Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Alessandro Marchioni
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
- Respiratory Disease Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Matteo Costantini
- Pathology Institute, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Luca Fabbiani
- Pathology Institute, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
- Immunohistochemistry Lab, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Filippo Genovese
- Centro Interdipartimentale Grandi Strumenti (C.I.G.S.), University of Modena and Reggio Emilia, Modena, Italy
| | - Diego Pinetti
- Centro Interdipartimentale Grandi Strumenti (C.I.G.S.), University of Modena and Reggio Emilia, Modena, Italy
| | - Linda Manicardi
- Respiratory Disease Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Ivana Castaniere
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
- Respiratory Disease Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Valentina Masciale
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italy
- Oncology Unit, University Hospital of Modena and Reggio Emilia, University of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Aramini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences-Diagnostic and Specialty Medicine (DIMEC) of the Alma Mater Studiorum, University of Bologna G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy
| | - Luca Tabbì
- Respiratory Disease Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Simone Rizzato
- Respiratory Disease Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Stefania Bettelli
- Molecular Pathology and Predictive Medicine Unit, Modena Cancer Center, University Hospital of Modena, Modena, Italy
| | - Samantha Manfredini
- Molecular Pathology and Predictive Medicine Unit, Modena Cancer Center, University Hospital of Modena, Modena, Italy
| | - Massimo Dominici
- Laboratory of Cellular Therapy, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, Modena, Italy
- Oncology Unit, University Hospital of Modena and Reggio Emilia, University of Modena and Reggio Emilia, Modena, Italy
| | - Enrico Clini
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
- Respiratory Disease Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Stefania Cerri
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
- Respiratory Disease Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| |
Collapse
|
42
|
Gavito-Covarrubias D, Ramírez-Díaz I, Guzmán-Linares J, Limón ID, Manuel-Sánchez DM, Molina-Herrera A, Coral-García MÁ, Anastasio E, Anaya-Hernández A, López-Salazar P, Juárez-Díaz G, Martínez-Juárez J, Torres-Jácome J, Albarado-Ibáñez A, Martínez-Laguna Y, Morán C, Rubio K. Epigenetic mechanisms of particulate matter exposure: air pollution and hazards on human health. Front Genet 2024; 14:1306600. [PMID: 38299096 PMCID: PMC10829887 DOI: 10.3389/fgene.2023.1306600] [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: 10/04/2023] [Accepted: 11/20/2023] [Indexed: 02/02/2024] Open
Abstract
Environmental pollution nowadays has not only a direct correlation with human health changes but a direct social impact. Epidemiological studies have evidenced the increased damage to human health on a daily basis because of damage to the ecological niche. Rapid urban growth and industrialized societies importantly compromise air quality, which can be assessed by a notable accumulation of air pollutants in both the gas and the particle phases. Of them, particulate matter (PM) represents a highly complex mixture of organic and inorganic compounds of the most variable size, composition, and origin. PM being one of the most complex environmental pollutants, its accumulation also varies in a temporal and spatial manner, which challenges current analytical techniques used to investigate PM interactions. Nevertheless, the characterization of the chemical composition of PM is a reliable indicator of the composition of the atmosphere, the quality of breathed air in urbanized societies, industrial zones and consequently gives support for pertinent measures to avoid serious health damage. Epigenomic damage is one of the most promising biological mechanisms of air pollution-derived carcinogenesis. Therefore, this review aims to highlight the implication of PM exposure in diverse molecular mechanisms driving human diseases by altered epigenetic regulation. The presented findings in the context of pan-organic cancer, fibrosis, neurodegeneration and metabolic diseases may provide valuable insights into the toxicity effects of PM components at the epigenomic level and may serve as biomarkers of early detection for novel targeted therapies.
Collapse
Affiliation(s)
- Dulcemaría Gavito-Covarrubias
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Ivonne Ramírez-Díaz
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
- Universidad Popular Autónoma del Estado de Puebla (UPAEP), Puebla, Mexico
| | - Josué Guzmán-Linares
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Ilhuicamina Daniel Limón
- Laboratory of Neuropharmacology, Faculty of Chemical Sciences, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Dulce María Manuel-Sánchez
- Laboratory of Neuropharmacology, Faculty of Chemical Sciences, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Alejandro Molina-Herrera
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Miguel Ángel Coral-García
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Estela Anastasio
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Arely Anaya-Hernández
- Centro de Investigación en Genética y Ambiente, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Primavera López-Salazar
- Centro de Investigaciones en Dispositivos Semiconductores (CIDS), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Gabriel Juárez-Díaz
- Centro de Investigaciones en Dispositivos Semiconductores (CIDS), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Javier Martínez-Juárez
- Centro de Investigaciones en Dispositivos Semiconductores (CIDS), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Julián Torres-Jácome
- Laboratorio de Fisiopatología Cardiovascular, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Alondra Albarado-Ibáñez
- Laboratorio de Fisiopatología Cardiovascular, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Ygnacio Martínez-Laguna
- Vicerrectoría de Investigación y Estudios de Posgrado, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Carolina Morán
- Centro de Investigación en Fisicoquímica de Materiales, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Karla Rubio
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| |
Collapse
|
43
|
Lan D, Fermoyle CC, Troy LK, Knibbs LD, Corte TJ. The impact of air pollution on interstitial lung disease: a systematic review and meta-analysis. Front Med (Lausanne) 2024; 10:1321038. [PMID: 38298511 PMCID: PMC10827982 DOI: 10.3389/fmed.2023.1321038] [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: 10/13/2023] [Accepted: 12/27/2023] [Indexed: 02/02/2024] Open
Abstract
Introduction There is a growing body of evidence suggesting a causal relationship between interstitial lung disease (ILD) and air pollution, both for the development of the disease, and driving disease progression. We aim to provide a comprehensive literature review of the association between air pollution, and ILD, including idiopathic pulmonary fibrosis (IPF). Methods We systematically searched from six online database. Two independent authors (DL and CF) selected studies and critically appraised the risk of bias using the Newcastle-Ottawa Scale (NOS). Findings are presented through a narrative synthesis and meta-analysis. Meta-analyses were performed exclusively when there was a minimum of three studies examining identical pollutant-health outcome pairs, all evaluating equivalent increments in pollutant concentration, using a random effects model. Results 24 observational studies conducted in 13 countries or regions were identified. Pollutants under investigation encompassed ozone (O3), nitrogen dioxide (NO2), Particulate matter with diameters of 10 micrometers or less (PM10) and 2.5 micrometers or less (PM2.5), sulfur dioxide (SO2), carbon monoxide (CO), nitric oxide (NO) and nitrogen oxides (NOx). We conducted meta-analyses to assess the estimated Risk Ratios (RRs) for acute exacerbations (AE)-IPF in relation to exposure to every 10 μg/m3 increment in air pollutant concentrations, including O3, NO2, PM10, and PM2.5. The meta-analysis revealed a significant association between the increased risk of AE-IPF in PM2.5, yielding RR 1.94 (95% CI 1.30-2.90; p = 0.001). Findings across all the included studies suggest that increased exposure to air pollutants may be linked to a range of health issues in individuals with ILDs. Conclusion A scarcity of available studies on the air pollutants and ILD relationship underscores the imperative for further comprehensive research in this domain. The available data suggest that reducing levels of PM2.5 in the atmosphere could potentially reduce AE frequency and severity in ILD patients.
Collapse
Affiliation(s)
- Doris Lan
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- National Health and Medical Research Council (NHMRC), Centre of Research Excellence in Pulmonary Fibrosis, Camperdown, NSW, Australia
| | - Caitlin C. Fermoyle
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- National Health and Medical Research Council (NHMRC), Centre of Research Excellence in Pulmonary Fibrosis, Camperdown, NSW, Australia
| | - Lauren K. Troy
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- National Health and Medical Research Council (NHMRC), Centre of Research Excellence in Pulmonary Fibrosis, Camperdown, NSW, Australia
| | - Luke D. Knibbs
- National Health and Medical Research Council (NHMRC), Centre of Research Excellence in Pulmonary Fibrosis, Camperdown, NSW, Australia
- School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Public Health Unit, Public Health Research Analytics and Methods for Evidence (PHRAME), Sydney Local Health District, Camperdown, NSW, Australia
| | - Tamera J. Corte
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Respiratory and Sleep Medicine, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- National Health and Medical Research Council (NHMRC), Centre of Research Excellence in Pulmonary Fibrosis, Camperdown, NSW, Australia
| |
Collapse
|
44
|
Zhang S, Hu W, Lv C, Song X. Biogenesis and Function of circRNAs in Pulmonary Fibrosis. Curr Gene Ther 2024; 24:395-409. [PMID: 39005062 DOI: 10.2174/0115665232284076240207073542] [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: 09/25/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 07/16/2024]
Abstract
Pulmonary fibrosis is a class of fibrosing interstitial lung diseases caused by many pathogenic factors inside and outside the lung, with unknown mechanisms and without effective treatment. Therefore, a comprehensive understanding of the molecular mechanism implicated in pulmonary fibrosis pathogenesis is urgently needed to develop new and effective measures. Although circRNAs have been widely acknowledged as new contributors to the occurrence and development of diseases, only a small number of circRNAs have been functionally characterized in pulmonary fibrosis. Here, we systematically review the biogenesis and functions of circRNAs and focus on how circRNAs participate in pulmonary fibrogenesis by influencing various cell fates. Meanwhile, we analyze the current exploration of circRNAs as a diagnostic biomarker, vaccine, and therapeutic target in pulmonary fibrosis and objectively discuss the challenges of circRNA- based therapy for pulmonary fibrosis. We hope that the review of the implication of circRNAs will provide new insights into the development circRNA-based approaches to treat pulmonary fibrosis.
Collapse
Affiliation(s)
- Songzi Zhang
- Department of Cellular and Genetic Medicine, Binzhou Medical University, Yantai, 264003, China
| | - Wenjie Hu
- Department of Cellular and Genetic Medicine, Binzhou Medical University, Yantai, 264003, China
| | - Changjun Lv
- Department of Cellular and Genetic Medicine, Binzhou Medical University, Yantai, 264003, China
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou 256603, China
| | - Xiaodong Song
- Department of Cellular and Genetic Medicine, Binzhou Medical University, Yantai, 264003, China
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou 256603, China
| |
Collapse
|
45
|
Luo W, Gu Y, Fu S, Wang J, Zhang J, Wang Y. Emerging opportunities to treat idiopathic pulmonary fibrosis: Design, discovery, and optimizations of small-molecule drugs targeting fibrogenic pathways. Eur J Med Chem 2023; 260:115762. [PMID: 37683364 DOI: 10.1016/j.ejmech.2023.115762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/15/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common fibrotic form of idiopathic diffuse lung disease. Due to limited treatment options, IPF patients suffer from poor survival. About ten years ago, Pirfenidone (Shionogi, 2008; InterMune, 2011) and Nintedanib (Boehringer Ingelheim, 2014) were approved, greatly changing the direction of IPF drug design. However, limited efficacy and side effects indicate that neither can reverse the process of IPF. With insights into the occurrence of IPF, novel targets and agents have been proposed, which have fundamentally changed the treatment of IPF. With the next-generation agents, targeting pro-fibrotic pathways in the epithelial-injury model offers a promising approach. Besides, several next-generation IPF drugs have entered phase II/III clinical trials with encouraging results. Due to the rising IPF treatment requirements, there is an urgent need to completely summarize the mechanisms, targets, problems, and drug design strategies over the past ten years. In this review, we summarize known mechanisms, target types, drug design, and novel technologies of IPF drug discovery, aiming to provide insights into the future development and clinical application of next-generation IPF drugs.
Collapse
Affiliation(s)
- Wenxin Luo
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yilin Gu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Siyu Fu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, Tennessee, United States
| | - Jifa Zhang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| |
Collapse
|
46
|
Tang L, Li D, Ma Y, Cui F, Wang J, Tian Y. The association between telomere length and non-alcoholic fatty liver disease: a prospective study. BMC Med 2023; 21:427. [PMID: 37940980 PMCID: PMC10634180 DOI: 10.1186/s12916-023-03136-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 10/27/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Research on the association between telomere length (TL) and incident non-alcoholic fatty liver disease (NAFLD) is limited. This study examined this association and further assessed how TL contributes to the association of NAFLD with its known risk factors. METHODS Quantitative PCR (polymerase chain reaction) was employed to assess leucocyte telomere length. Polygenic risk score (PRS) for NAFLD, air pollution score, and lifestyle index were constructed. Cox proportional hazard models were conducted to estimate the hazard ratios (HRs) and 95% confidence intervals. RESULTS Among 467,848 participants in UK Biobank, we identified 4809 NAFLD cases over a median follow-up of 12.83 years. We found that long TL was associated with decreased risk of incident NAFLD, as each interquartile range increase in TL resulted in an HR of 0.93 (95% CI 0.89, 0.96). TL partly mediated the association between age and NAFLD (proportion mediated: 15.52%). When assessing the joint effects of TL and other risk factors, the highest risk of NAFLD was found in participants with low TL and old age, low TL and high air pollution score, low TL and unfavorable lifestyle, and low TL and high PRS, compared to each reference group. A positive addictive interaction was observed between high PRS and low TL, accounting for 14.57% (2.51%, 27.14%) of the risk of NAFLD in participants with low telomere length and high genetic susceptibility. CONCLUSIONS Long telomere length was associated with decreased risk of NAFLD incidence. Telomere length played an important role in NAFLD.
Collapse
Affiliation(s)
- Linxi Tang
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China
| | - Dankang Li
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China
| | - Yudiyang Ma
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China
| | - Feipeng Cui
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China
| | - Jianing Wang
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China
| | - Yaohua Tian
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China.
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No.13 Hangkong Road, Wuhan, 430030, China.
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences/Peking Union Medical College, No.31, Beijige-3, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
47
|
Goobie GC. Where you live matters: Roadways, air pollution and lung function in patients with idiopathic pulmonary fibrosis. Respirology 2023; 28:906-908. [PMID: 37536709 DOI: 10.1111/resp.14565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/05/2023]
Abstract
See related article
Collapse
Affiliation(s)
- Gillian C Goobie
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
48
|
Ma Y, Cui F, Li D, Wang J, Tang L, Xie J, Hu Y, Tian Y. Lifestyle, Genetic Susceptibility, and the Risk of Idiopathic Pulmonary Fibrosis: A Large Prospective Cohort Study. Chest 2023; 164:929-938. [PMID: 37059176 DOI: 10.1016/j.chest.2023.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/28/2023] [Accepted: 04/05/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND Lifestyle is an important contributor of age-related chronic disease, but the association between lifestyle and the risk of idiopathic pulmonary fibrosis (IPF) remains unknown. The extent to which genetic susceptibility modifies the effects of lifestyle on IPF also remains unclear. RESEARCH QUESTION Is there a joint effect or interaction of lifestyle and genetic susceptibility on the risk of developing IPF? STUDY DESIGN AND METHODS This study included 407,615 participants from the UK Biobank study. A lifestyle score and a polygenic risk score were constructed separately for each participant. Participants were then classified into three lifestyle categories and three genetic risk categories based on the corresponding score. Cox models were fitted to assess the association of lifestyle and genetic risk with the risk of incident IPF. RESULTS With favorable lifestyle as the reference group, intermediate lifestyle (hazard ratio, 1.384; 95% CI, 1.218-1.574) and unfavorable lifestyle (hazard ratio, 2.271; 95% CI, 1.852-2.785) were significantly associated with an increased risk of IPF. For the combined effect of lifestyle and polygenic risk score, participants with unfavorable lifestyle and high genetic risk had the highest risk of IPF (hazard ratio, 7.796; 95% CI, 5.482-11.086) compared with those with favorable lifestyle and low genetic risk. Moreover, approximately 32.7% (95% CI, 11.3-54.1) of IPF risk could be attributed to the interaction of an unfavorable lifestyle and high genetic risk. INTERPRETATION Exposure to unfavorable lifestyle significantly increased the risk of IPF, particularly in those with high genetic risk.
Collapse
Affiliation(s)
- Yudiyang Ma
- Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feipeng Cui
- Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dankang Li
- Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianing Wang
- Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linxi Tang
- Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junqing Xie
- Center for Statistics in Medicine, NDORMS, University of Oxford, The Botnar Research Centre, Oxford, UK
| | - Yonghua Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Yaohua Tian
- Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
49
|
Abstract
PURPOSE OF REVIEW Genetics contributes substantially to the susceptibility to idiopathic pulmonary fibrosis (IPF). Genetic studies in sporadic and familial disease have identified several IPF-associated variants, mainly in telomere-related and surfactant protein genes.Here, we review the most recent literature on genetics of IPF and discuss how it may contribute to disease pathogenesis. RECENT FINDINGS Recent studies implicate genes involved in telomere maintenance, host defence, cell growth, mammalian target of rapamycin signalling, cell-cell adhesion, regulation of TGF-β signalling and spindle assembly as biological processes involved in the pathogenesis of IPF. Both common and rare genetic variants contribute to the overall risk of IPF; however, while common variants (i.e. polymorphisms) account for most of the heritability of sporadic disease, rare variants (i.e. mutations), mainly in telomere-related genes, are the main contributors to the heritability of familial disease. Genetic factors are likely to also influence disease behaviour and prognosis. Finally, recent data suggest that IPF shares genetic associations - and probably some pathogenetic mechanisms - with other fibrotic lung diseases. SUMMARY Common and rare genetic variants are associated with susceptibility and prognosis of IPF. However, many of the reported variants fall in noncoding regions of the genome and their relevance to disease pathobiology remains to be elucidated.
Collapse
Affiliation(s)
- Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Joyce S Lee
- University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
50
|
Stanel SC, Callum J, Rivera-Ortega P. Genetic and environmental factors in interstitial lung diseases: current and future perspectives on early diagnosis of high-risk cohorts. Front Med (Lausanne) 2023; 10:1232655. [PMID: 37601795 PMCID: PMC10435297 DOI: 10.3389/fmed.2023.1232655] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
Within the wide scope of interstitial lung diseases (ILDs), familial pulmonary fibrosis (FPF) is being increasingly recognized as a specific entity, with earlier onset, faster progression, and suboptimal responses to immunosuppression. FPF is linked to heritable pathogenic variants in telomere-related genes (TRGs), surfactant-related genes (SRGs), telomere shortening (TS), and early cellular senescence. Telomere abnormalities have also been identified in some sporadic cases of fibrotic ILD. Air pollution and other environmental exposures carry additive risk to genetic predisposition in pulmonary fibrosis. We provide a perspective on how these features impact on screening strategies for relatives of FPF patients, interstitial lung abnormalities, ILD multi-disciplinary team (MDT) discussion, and disparities and barriers to genomic testing. We also describe our experience with establishing a familial interstitial pneumonia (FIP) clinic and provide guidance on how to identify patients with telomere dysfunction who would benefit most from genomic testing.
Collapse
Affiliation(s)
- Stefan Cristian Stanel
- Interstitial Lung Disease Unit, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Jack Callum
- Interstitial Lung Disease Unit, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Pilar Rivera-Ortega
- Interstitial Lung Disease Unit, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|